WO2020006374A2 - Anticorps anti-sirp-bêta1 et procédés d'utilisation associés - Google Patents

Anticorps anti-sirp-bêta1 et procédés d'utilisation associés Download PDF

Info

Publication number
WO2020006374A2
WO2020006374A2 PCT/US2019/039757 US2019039757W WO2020006374A2 WO 2020006374 A2 WO2020006374 A2 WO 2020006374A2 US 2019039757 W US2019039757 W US 2019039757W WO 2020006374 A2 WO2020006374 A2 WO 2020006374A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
amino acid
hvr
acid sequence
seq
Prior art date
Application number
PCT/US2019/039757
Other languages
English (en)
Other versions
WO2020006374A3 (fr
Inventor
Andrew PINCETIC
Patricia Culp
Arnon Rosenthal
Original Assignee
Alector Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP19744960.6A priority Critical patent/EP3814377A2/fr
Priority to AU2019293589A priority patent/AU2019293589A1/en
Priority to KR1020217002521A priority patent/KR20210025614A/ko
Priority to CN201980040821.9A priority patent/CN112384532A/zh
Priority to CA3099176A priority patent/CA3099176A1/fr
Priority to US17/256,508 priority patent/US20210277113A1/en
Priority to JP2020572914A priority patent/JP2021529520A/ja
Priority to MX2020013324A priority patent/MX2020013324A/es
Application filed by Alector Llc filed Critical Alector Llc
Priority to EA202190138A priority patent/EA202190138A1/ru
Priority to SG11202010990TA priority patent/SG11202010990TA/en
Priority to BR112020026819-4A priority patent/BR112020026819A2/pt
Publication of WO2020006374A2 publication Critical patent/WO2020006374A2/fr
Publication of WO2020006374A3 publication Critical patent/WO2020006374A3/fr
Priority to IL279648A priority patent/IL279648A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present disclosure relates to anti-SIRP 1 antibodies and therapeutic uses of such antibodies.
  • SIRP Signal regulatory protein beta
  • the SIRP family of proteins is characterized by an extracellular region containing 2 membrane-proximal IgC domains and a distal IgV domain.
  • SIRP proteins contain short cytoplasmic domains that lack cytoplasmic sequence motifs capable of recruiting protein tyrosine phosphatase SHP-2 and SHP-l.
  • SIRP i isoform 1 associates with the adaptor protein DAP12, which contains a single cytoplasmic immunoreceptor tyrosine-based activating (ITAM) motif.
  • ITAM immunoreceptor tyrosine-based activating
  • SIRP has been shown to be a microglial modulator of phagocytosis in Alzheimer’s disease. See, e.g., Gaikwad et at. 2009 Am J Pathol 175:2528-2539.
  • an isolated antibody that binds to human SIRP 1 wherein the antibody has at least one, at least two at least three, at least four, or at least five properties selected from:
  • g induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro and/or in vivo;
  • h) induces or increases IL-8 expression in monocytes in vitro and/or in vivo; i) induces or increases TNFa expression in macrophages and/or dendritic cells in vitro and/or in vivo;
  • m increases viability of macrophages in vitro and/or in vivo, alone and/or in combination with an agonist anti-TREM2 antibody
  • n increases viability of dendritic cells in vitro and/or in vivo.
  • the antibody binds to human SIRP 1 isoform 1, but does not bind to human SIRPa or human SIRP 1 isoform 3. In some embodiments, the antibody binds to human SIRP 1 isoform 1, but does not bind to human SIRPa, SIRPy, or human SIRP 1 isoform 3. In some embodiments, the antibody has at least one, at least two, or at least three properties selected from: a) agonizes SIRPp i activity on CD 14-positive monocytes in vitro and/or in vivo;
  • e induces neutrophil-mediated phagocytosis, for example, of tumor cells in vitro and/or in vivo;
  • an isolated antibody that binds to human SIRPb 1 comprises (a) HVR-H1 comprising an amino acid sequence of an FTVR- Hl shown in Table 4 and/or Table 9; (b) HVR-H2 comprising an amino acid sequence of an FTVR- H2 shown in Table 4 and/or Table 9; (c) HVR-H3 comprising an amino acid sequence of an FTVR- H3 shown in Table 4 and/or Table 9; (d) HVR-L1 comprising an amino acid sequence of an FTVR- Ll shown in Table 3 and/or Table 8; (e) HVR-L2 comprising an amino acid sequence of an HVR-L2 shown in Table 3 and/or Table 8; and (f) HVR-L3 comprising an amino acid sequence of an FTVR- L3 shown in Table 3 and/or Table 8.
  • the heavy chain variable region comprises one, two, three or four framework regions selected from VH FR1, VH FR2, VH FR3, and VH FR4 shown in Table 6.
  • the light chain variable region comprises one, two, three or four framework regions selected from VL FR1, VL FR2, VL FR3, and VL FR4 shown in Table 5.
  • the antibody comprises HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB-17, SB-18, SB-19, SB-20, SB- 21, SB-22, SB-23, SB-24, SB-25, SB-26, SB-27, SB-28, SB-29, SB-30, SB-31, SB-32, SB-33, SB- 34, SB-35, SB-36, SB-37, SB-38, SB-39, SB-40, SB-41, SB-42, SB-43, SB-44, SB-45, SB-46, SB-
  • the antibody comprises a heavy chain variable region comprising an amino acid sequence at least 90%, at least 95%, at least 97%, or at least 99% identical to a sequence selected from SEQ ID NOs: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325,
  • the antibody comprises a heavy chain variable region selected from SEQ ID NOs: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307,
  • the antibody comprises a light chain variable region comprising an amino acid sequence at least 90%, at least 95%, at least 97%, or at least 99% identical to a sequence selected from SEQ ID NOs: 267, 269, 271, 273, 275, 277, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302, 304, 306, 308, 310, 312, 314, 316, 318, 320, 322,
  • the antibody comprises a light chain variable region selected from SEQ ID NOs: 267, 269, 271, 273, 275, 277, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296,
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the heavy chain variable region, and a light chain variable region 90%, at least 95%, at least 97%, or at least 99% identical to the light chain variable region, of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-
  • SB-40 SB-41, SB-42, SB-43, SB-44, SB-45, SB-46, SB-47, SB-48, SB-49, SB-50, SB-l-2, SB- 1-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB- 8-16, SB-40- 18, SB-40- 19, SB-40-20, and SB-40-21.
  • the antibody comprises a heavy chain variable region and a light chain variable region of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB- 16, SB-17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-24, SB-25, SB-26, SB-27, SB-28, SB- 29, SB-30, SB-31, SB-32, SB-33, SB-34, SB-35, SB-36, SB-37, SB-38, SB-39, SB-40, SB-41, SB- 42, SB-43, SB-44, SB-45, SB-46, SB-47, SB-48, SB-49, SB-50, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-l, SB
  • the antibody comprises HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3, 4, 8, and 9).
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the heavy chain variable region of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-
  • the antibody comprises a heavy chain variable region of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the light chain variable region of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a light chain variable region of an antibody selected from SB-l, SB-2, SB-3, SB- 4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40- 18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the heavy chain variable region, and a light chain variable region 90%, at least 95%, at least 97%, or at least 99% identical to the light chain variable region, of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l- 4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB- 40-18, SB-40- 19, SB-40-20, and SB-40-21.
  • the antibody comprises a heavy chain variable region and a light chain variable region of an antibody selected from SB-l, SB-2, SB-
  • the antibody comprises HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the heavy chain variable region of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB- 40-20, and SB-40-21.
  • the antibody comprises a heavy chain variable region of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the light chain variable region of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8- 14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a light chain variable region of an antibody selected from SB-l-2, SB-l-3, SB-l-
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the heavy chain variable region, and a light chain variable region 90%, at least 95%, at least 97%, or at least 99% identical to the light chain variable region, of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises a heavy chain variable region and a light chain variable region of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 80, 228, or 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 101, 239, 239, 240, or 241; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 125; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 366, 367, 368, or 369. In some embodiments, the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 267. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 366, 367, 368, or 369. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 267.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 81, 99, 230, 231, or 232; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 102, 242, 243, 244, or 245; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 126 or 253; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 371, 372, 373, 374, or 375. In some embodiments, the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 269 or 370. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 371, 372, 373, 374, or 375. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 269 or 370.
  • the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 371, 372, or 373 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 370; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 374 or 375 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 269.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 85, 233, or 234; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 107, 246, 247, or 248; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 131 or 254; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 376, 377, or 378. In some embodiments, the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 280. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 376, 377, or 378. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 280.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 97, 235, 236, or 237; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 121, 249, 250, 251, or 252; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 163; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 2; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 22; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 379, 380, 381, or 382. In some embodiments, the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 344. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 379, 380, 381, or 382. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 344.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 367.
  • the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 267. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 367. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 267.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 231 ; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 372.
  • the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 370. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 372. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 270.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO:
  • the antibody comprises a heavy chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 376.
  • the antibody comprises a light chain variable region that is at least 90%, at least 95%, at least 97%, or at least 99% identical to the amino acid sequence of SEQ ID NO: 280. In some embodiments, the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 376. In some embodiments, the antibody comprises a light chain variable region comprising the amino acid sequence of SEQ ID NO: 280.
  • the antibody is a monoclonal antibody. In some embodiments, the antibody is of the IgG class, the IgM class, or the IgA class. In some embodiments, the antibody is of the IgG class and has an IgGl, IgG2, or IgG4 isotype. In some embodiments, the antibody has an IgGl isotype. In some embodiments, the antibody comprises a E430G substitution and a P331 S substitution according to EU numbering.
  • the antibody is an antibody fragment.
  • the fragment is a Fab, Fab’, Fab’-SH, F(ab’)2, Fv or scFv fragment.
  • the antibody has an affinity (KD) for human SIRP 1 isoform 1 of 0.1 nM to 50 nM, or 0.5 nM to 10 nM, or 0.5 nM to 5 nM.
  • affinity is measured using a ForteBio Octet ® system.
  • the antibody recognizes a first and a second antigen, wherein the first antigen is SIRPpl and the second antigen is:
  • an antigen facilitating transport across the blood-brain-barrier selected from the group consisting of transferrin receptor (TR), insulin receptor (HIR), insulin-like growth factor receptor (IGFR), low-density lipoprotein receptor related proteins 1 and 2 (LPR-l and 2), and diphtheria toxin receptor;
  • the disease-causing nucleic acids are antisense GGCCCC (G2C4) repeat-expansion RNA
  • the disease-causing proteins are selected from the group consisting of amyloid beta, oligomeric amyloid beta, amyloid beta plaques, amyloid precursor protein or fragments thereof, Tau, IAPP, alpha-synuclein, TDP-43, FUS protein, C9orf72 (chromosome 9 open reading frame 72), c9RAN protein, prion protein, PrPSc, huntingtin, calcitonin, superoxide dismutase, ataxin, ataxin 1, ataxin 2, ataxin 3, ataxin 7, ataxin 8, ataxin 10, Lewy body, atrial natriuretic factor, islet amyloid polypeptide, insulin, apolipoprotein AI, serum amyloid A, medin, prolactin, transthyretin, lysozyme, beta 2
  • an isolated antibody that binds to human SIRP 1 is provided, wherein the antibody competes with one or more antibodies provided herein for binding to human SIRP 1 isoform 1.
  • an isolated antibody that binds to human SIRP 1 is provided, wherein the antibody binds essentially the same or overlapping SIRP 1 isoform 1 epitope as one or more antibodies provided herein.
  • an isolated nucleic acid comprising a nucleic acid sequence encoding an anti-SIRP 1 antibody provided herein.
  • a vector comprising the nucleic acid is provided.
  • an isolated host cell is provided, comprising the vector.
  • an isolated host cell is provided that expresses an anti-SIRP 1 antibody provided herein.
  • methods of producing an antibody that binds to human SIRP 1 comprising culturing a host cell that expresses an anti- SIRP 1 antibody provided herein so that the antibody is produced.
  • the method further comprises recovering the antibody produced by the cell.
  • a pharmaceutical composition comprising an anti- SIRP 1 antibody provided herein and a pharmaceutically acceptable carrier.
  • a method of treating cancer comprising administering to an individual in need thereof a therapeutically effective amount of an anti-SIRP 1 antibody provided herein.
  • the cancer is selected from selected from sarcoma, bladder cancer, brain cancer, breast cancer, colon cancer, rectal cancer, endometrial cancer, kidney cancer, renal pelvis cancer, leukemia, lung cancer, small cell lung cancer, melanoma, lymphoma, pancreatic cancer, prostate cancer, ovarian cancer, and fibrosarcoma, glioblastoma multiforme; renal clear cell carcinoma; adrenocortical carcinoma; bladder urothelial carcinoma, diffuse large B-cell lymphoma, lung adenocarcinoma; pancreatic adenocarcinoma, renal cell cancer, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, indolent B cell lymphoma, aggressive B cell lymphoma, T cell lymph
  • adenocarcinoma diffuse large B-cell lymphoma, esophageal carcinoma, head and neck squamous cell carcinoma, kidney chromophobe, renal papillary cell carcinoma, lower grade glioma, hepatocellular carcinoma, lung squamous cell carcinoa, mesothelioma, ovarian serous cystadenomcarcinoma, pancreatic adenocarcinoma, pheochromocytoma and paraganglioma, prostate adenocarconimo, rectal adenocarcinoma, cutaneous melanoma, stomach
  • adenocarcinoma testicular germ cell tumors, thyroid carcinoma, thyumoma, uterine corpus endometrial carcinoma, uternine carcinosarcoma, and uveal melanoma.
  • a method of treatment further comprises administering a therapeutic agent that inhibits or agonizes PD1, PDL1, CD40, 0X40, ICOS, CD28, CD137/4-1BB, CD27, GITR, CTLA4, PD-L2, B7-H3, B7-H4, HVEM, LIGHT, BTLA, CD30, TIGIT, VISTA, KIR, GAL9, TIM1, TIM3, TIM4, A2AR, LAG3, DR-5, CD2, CD5, CD39, or CD73.
  • a method of treatment further comprises administering to the individual at least one antibody that specifically binds to an inhibitory checkpoint molecule, and/or one or more standard or investigational anti-cancer therapies.
  • a method of treatment further comprises administering at least one antibody that specifically binds to an inhibitory checkpoint molecule in combination with the anti-SIRPA antibody.
  • the at least one antibody that specifically binds to an inhibitory checkpoint molecule is selected from an anti-PD-Ll antibody, an anti-CTLA4 antibody, an anti-PD-L2 antibody, an anti -PD- 1 antibody, an anti-B7-H3 antibody, an anti-B7- H4 antibody, and anti-HVEM antibody, an anti- B- and T-lymphocyte attenuator (BTLA) antibody, an anti-Killer inhibitory receptor (KIR) antibody, an anti-GAL9 antibody, an anti- TIM-l antibody, an anti-TIM3 antibody, an anti-TIM-4 antibody, an anti-A2AR antibody, an anti-CD39 antibody, an anti-CD73 antibody, an anti -LAG-3 antibody, an anti
  • phosphatidyl serine antibody an anti-CD27 antibody, an anti-CD30 antibody, an anti-TNFa antibody, an anti-CD33 antibody, an anti-Siglec-5 antibody, an anti-Siglec-7 antibody, an anti- Siglec-9 antibody, an anti-Siglec-l l antibody, an antagonistic anti-TREMl antibody, an antagonistic anti-TREM2 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti- CD2 antibody, an anti-CD5 antibody, and any combination thereof.
  • a method of treatment further comprises administering one or more standard or investigational anti-cancer therapies selected from radiotherapy, cytotoxic chemotherapy, targeted therapy, imatinib therapy, trastuzumab therapy, etanercept therapy, adoptive cell transfer (ACT) therapy, chimeric antigen receptor T cell transfer (CAR-T) therapy, vaccine therapy, and cytokine therapy.
  • standard or investigational anti-cancer therapies selected from radiotherapy, cytotoxic chemotherapy, targeted therapy, imatinib therapy, trastuzumab therapy, etanercept therapy, adoptive cell transfer (ACT) therapy, chimeric antigen receptor T cell transfer (CAR-T) therapy, vaccine therapy, and cytokine therapy.
  • a method of treatment further comprises administering to the individual at least one antibody that specifically binds to an inhibitory cytokine.
  • the at least one antibody that specifically binds to an inhibitory cytokine is selected from an anti-CCL2 antibody, an anti-CSF-l antibody, an anti-IL-2 antibody, and any combination thereof.
  • a method of treatment further comprises administering to the individual at least one agonistic antibody that specifically binds to a stimulatory checkpoint protein.
  • the at least one agonistic antibody that specifically binds to a stimulatory checkpoint protein is selected from an agonist anti-CD40 antibody, an agonist anti- 0X40 antibody, an agonist anti-ICOS antibody, an agonist anti-CD28 antibody, an agonistic anti-TREMl antibody, an agonistic anti-TREM2 antibody, an agonist anti-CDl37/4-lBB antibody, an agonist anti-CD27 antibody, an agonist anti-glucocorticoid-induced TNFR-related protein GITR antibody, an agonist anti-CD30 antibody, an agonist anti-BTLA antibody, an agonist anti-HVEM antibody, an agonist anti-CD2 antibody, an agonist anti-CD5 antibody, and any combination thereof.
  • a method of treatment further comprises administering to the individual at least one stimulatory cytokine.
  • the at least one stimulatory cytokine is selected from IFN-a4, IFN-b, IL- 1 b, TNF-a, IL-6, IL-8, CRP, IL-20 family members, LIF, IFN-g, OSM, CNTF, GM-CSF, IL-l l, IL-12, IL-15, IL-17, IL-18, IL-23, CXCL10, IL-33, MCP-l, MIP-l-beta, and any combination thereof.
  • an anti-SII ⁇ l antibody provided herein for the preparation of a medicament.
  • the medicament is for treating cancer.
  • an anti-SII ⁇ l antibody provided herein is provided for treating cancer.
  • a method of treating a neurodegenerative disease or disorder comprising administering to an individual in need thereof a therapeutically effective amount of an anti-SIiy ⁇ l antibody provided herein.
  • the neurodegenerative disease or disorder is selected from dementia, frontotemporal dementia, progressive
  • Alzheimer’s disease vascular dementia, Nasu-Hakola disease, cognitive deficit, memory loss, seizures, retinal dystrophy, amyotrophic lateral sclerosis, traumatic brain injury, a spinal cord injury, stroke, Parkinson’s disease, acute disseminated encephalomyelitis, retinal degeneration, age related macular degeneration, glaucoma, multiple sclerosis.
  • an anti-SIRP 1 antibody provided herein for the preparation of a medicament for treating a neurodegenerative disease or disorder.
  • the neurodegenerative disease or disorder is selected from dementia,
  • an anti-SIRP 1 antibody provided herein for use in treating a neurodegenerative disease or disorder is provided.
  • the neurodegenerative disease or disorder is selected from dementia,
  • frontotemporal dementia progressive supranuclear palsy, Alzheimer’s disease, vascular dementia, Nasu-Hakola disease, cognitive deficit, memory loss, seizures, retinal dystrophy, amyotrophic lateral sclerosis, traumatic brain injury, a spinal cord injury, stroke, Parkinson’s disease, acute disseminated encephalomyelitis, retinal degeneration, age related macular degeneration, glaucoma, multiple sclerosis.
  • FIG. 1A shows an alignment of the amino acid sequences of SIRP 1 isoform 1 (SEQ ID NO: 1; Uniprot Accession No. 000241) and SIRP l isoform 3 (SEQ ID NO: 384; Uniprot Accession No. Q5TFQ8).
  • FIG. IB shows an alignment of the amino acid sequences of SIRP 1 isoform 1 (SEQ ID NO: 1) with SIRPa (SEQ ID NO: 385; Uniprot Accession No. P78324), which shows high homology within the extracellular domains.
  • FIG. 2 shows an alignment of the amino acid sequences of human SIRP 1 isoform 1 (SEQ ID NO: 1) and mouse SIRP l (SEQ ID NO: 386; Uniprot Accession No. Q8BFX8).
  • FIG. 3A-3B shows induction of human SIRPpl -dependent luciferase expression in a cell- based reporter assay.
  • Cells were stimulated with plate-bound anti-SIRP 1 antibodies (3 A) or anti- SIRPa (SA-9C2) (3B) or human IgGl isotype control. Results are expressed as fold over background. The background level is set to 1 on y-axis.
  • FIG. 4A shows the expression pattern of two DAPl2-associated receptors, SIRP 1 and TREM1, on human monocytes. Shaded histograms represent background fluorescence from isotype stained cells. Black outlined histograms represent receptor expression level with target-specific antibody stained cells.
  • FIG. 4B shows the expression of SIRP 1 on Ml and M2 polarized macrophages from 2 healthy donors. Shaded histograms represent background fluorescence from isotype stained cells. Black outlined histograms represent SIRP 1 expression level on M2 macrophages, whereas dashed line histograms represent SIRP 1 expression level on Ml
  • FIG. 4C shows the expression of SIRP 1 and TREM1 on human monocyte-derived dendritic cells (DCs) with or without LPS stimulation. Shaded histograms represent background fluorescence from isotype stained cells. Black outlined histograms represent receptor expression level with target-specific antibody stained cells.
  • DCs human monocyte-derived dendritic cells
  • FIG. 5A profiles the RNA expression pattern of SIRP 1 isoform 1 in various tissues from human donors.
  • FIG. 5B profiles the RNA expression pattern of SIRP 1 isoform 3 in various tissues from human donors. Peak expression for SIRP 1 isoform 3 occurs in the brain with limited expression in peripheral tissues.
  • FIG. 6A shows bIKRbI -mediated respiratory burst from primary human neutrophils obtained from two different donors.
  • FIG. 6B shows SIRP 1 -mediated respiratory burst (top panel) or IL-8 release (bottom panel) from primary human monocytes.
  • FIG. 6C shows SIRP 1 -mediated TNFa cytokine release from primary human macrophages or dendritic cells (DCs).
  • FIG. 7 shows the anti-tumor activity of primary human neutrophils stimulated with plate- bound anti-SIRP 1 antibodies. Luminescence values are presented on a relative scale with the signal of Raji-Luc co-cultured with neutrophils in the absence of opsonizing antibody set to 1 on the y-axis.
  • FIG. 8 shows induction of human SIRPpl -dependent luciferase expression in a cell-based reporter assay by affinity matured antibodies. Results presented are raw luminescence values. The background level is set to 10,000 on y-axis.
  • FIG. 9 shows SIRP l -mediated TNFa cytokine release from primary human dendritic cells (DCs).
  • FIG. 10A-10B shows the cross-reactivity of anti-SIRP 1 antibodies to multiple SIRP 1 antigens.
  • Soluble, human SIRPpl-Fc isoform 1 (000241) or isoform 3 (Q5TFQ8) or cynomolgus SIRP 1 -Fc (XM005568541) were coated onto plates and incubated with increasing concentrations of parental and affinity matured forms of anti-SIRP 1 antibodies SB-l, SB- 1-2, SB-2, SB-2-7, SB-8, SB-8-13, SB-40, and SB-40-21. All anti-SIRP l antibodies bound only to human SIRP l-Fc isoform 1 antigen.
  • FIG. 11A shows the upregulation of TREM2 expression on monocyte-derived human macrophages from three different healthy donors by plate-bound, full-length anti-SIRPP 1 antibodies SB-l-3, SB-2-8, SB-8-13, SB-8-15, and SB-40-20.
  • Baseline TREM2 expression was determined from cells incubated on human IgGl isotype control antibody.
  • TREM2 was detected with an anti-TREM2 antibody (clone ADI-22) conjugated with DyLight650
  • FIG. 11B shows the viability of monocyte-derived human macrophages cultured on plate-bound, full-length anti-SIRPpl antibodies SB-l-3, SB-2-8, SB-8-13, SB-8-15, and SB- 40-20. Viable cells were quantified by measuring luminescence values following addition of Cell Titer Glo substrate. Cells cultured on human IgGl isotype control or in the absence of plate-bound antibody (No Ab) established baseline viability of cells.
  • FIG. 12 shows the viability of monocyte-derived macrophages from two different healthy donors cultured on increasing concentrations of plate-bound, full-length anti-SIRPp i antibodies SB-l-3 and SB-2-8 or human IgGl isotype control. Viable cells were quantified by measuring luminescence values following addition of Cell Titer Glo substrate.
  • FIG. 13 shows anti-SIRPp i antibodies enhance the agonistic activity of anti-TREM2 antibody in a macrophage viability assay.
  • Monocyte-derived macrophages were cultured on plate-bound, full-length SB-l-3, SB-2-8, or huIgGl isotype control in the presence or absence of soluble anti-TREM2 antibody.
  • Viable cells were quantified by measuring luminescence values following addition of Cell Titer Glo substrate.
  • FIG. 14A shows the viability of bone marrow-derived macrophages obtained from human SIRPp i BAC transgenic mice cultured on plate-bound, full-length anti-SIRPp i antibodies SB-l-3, SB-2-8, and SB-8-13 or human IgGl isotype control.
  • FIG. 14B shows the viability of bone marrow-derived dendritic cells obtained from human SIRPp i BAC transgenic mice cultured on plate-bound, full-length anti-SIRPp i antibodies SB-l-3, SB-2-8, and SB-8-13 or human IgGl isotype control. Viable cells were quantified by measuring luminescence values following addition of Cell Titer Glo substrate. Cells cultured on human IgGl isotype control or in the absence of plate-bound antibody (No Ab) established baseline viability of cells.
  • FIG. 15A-15C show cross-reactivity of anti-SIRP 1 antibodies to different receptors of the SIRP family.
  • FIG. 15A and FIG. 15B show titration curves of various anti-SIRP receptor antibodies binding BW5147.G.1.4 cells overexpressing recombinant human SIRP 1 or recombinant human SIRPa, respectively.
  • FIG. 15C shows titration curves of antibodies binding Jurkat cells, an immortalized human T cell line that endogenously expresses SIRPy. Positive control antibodies were anti-SIRPa/b clone 18D5, anti-SIRPa/b/g clone KWAR23, and anti-SIRPy clone LSB2.20.
  • anti-SIiy ⁇ l antibodies e.g., monoclonal antibodies
  • methods of making and using such antibodies pharmaceutical compositions comprising such antibodies; nucleic acids encoding such antibodies; and host cells comprising nucleic acids encoding such antibodies.
  • the techniques and procedures described or referenced herein are generally well understood and commonly employed using conventional methodology by those skilled in the art, such as, for example, the widely utilized methodologies such as those described in Sambrook et al. Molecular Cloning: A Laboratory Manual 3d edition (2001) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Current Protocols in Molecular Biology (F.M. Ausubel, et al. eds., (2003);
  • SIRP 1 polypeptide refer herein to any native SIRP 1 from any vertebrate source, including mammals such as primates (e.g ., humans and cynos) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term encompasses both wild-type sequences and naturally occurring variant sequences, e.g., splice variants or allelic variants.
  • the term encompasses“full-length,” unprocessed SIRP 1 as well as any form of SIRP 1 that results from processing in the cell.
  • the SIRP 1 is human SIRP 1
  • the amino acid sequence of an exemplary SIRP 1 isoform 1 is Uniprot Accession No. 000241 as of February 28, 2018.
  • the amino acid sequence of an exemplary human SIRP 1 is SEQ ID NO: 1.
  • the amino acid sequence of an exemplary SIRP 1 isoform 3 is Uniprot Accession No. Q5TFQ8 as of January 31, 2018.
  • the amino acid sequence of an exemplary human SIRP 1 isoform 3 is SEQ ID NO: 384. Unless specifically indicated otherwise,“SIRPQ 1” as used herein refers to SIRP 1 isoform 1.
  • the terms“anti- SIRP 1 antibody,” an“antibody that binds to SIRP 1 ,” and“antibody that specifically binds SIRPQ 1” refer to an antibody that is capable of binding SIRP 1 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting SIRP 1.
  • the extent of binding of an anti- SIRP 1 antibody to an unrelated, non- SIRP 1 polypeptide is less than about 10% of the binding of the antibody to SIRP 1 as measured, e.g., by a radioimmunoassay (RIA).
  • an antibody that binds to SIRP 1 has a dissociation constant (KD) of ⁇ 1 mM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g., 10 8 M or less, e.g. from 10 8 M to 10 13 M, e.g., from 10 9 M to 10 13 M).
  • KD dissociation constant
  • an anti-SIRP 1 antibody binds to an epitope of SIRP 1 that is conserved among SIRP 1 polypeptides from different species.
  • the term“specific binding” or “specifically binds” or is“specific for” a particular polypeptide or an epitope on a particular polypeptide target means binding that is measurably different from a non-specific interaction.
  • Specific binding can be measured, for example, by determining binding of a molecule compared to binding of a control molecule. For example, specific binding can be determined by competition with a control molecule that is similar to the target, for example, an excess of non-labeled target. In this case, specific binding is indicated if the binding of the labeled target to a probe is competitively inhibited by excess unlabeled target.
  • telomere binding or“specifically binds to” or is “specific for” a particular polypeptide or an epitope on a particular polypeptide target as used herein can be exhibited, for example, by a molecule having a KD for the target of about any of 10 4 M or lower, 10 5 M or lower, 10 6 M or lower, 10 7 M or lower, 10 8 M or lower, 10 9 M or lower, 10 10 M or lower, 10 11 M or lower, 10 12 M or lower or a KD in the range of 10 4 M to 10 6 M or 10 6 M to 10 10 M or 10 7 M to 10 9 M.
  • affinity and KD values are inversely related.
  • the term“specific binding” refers to binding where a molecule binds to a particular polypeptide or epitope on a particular polypeptide without substantially binding to any other polypeptide or polypeptide epitope.
  • immunoglobulin (Ig) is used interchangeably with“antibody” herein.
  • antibody herein is used in the broadest sense and specially covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies ( e.g ., bispecific antibodies) including those formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • “Native antibodies” are usually heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical Light (“L”) chains and two identical heavy (“FI”) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intra-chain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains.
  • VH variable domain
  • Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
  • the light chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (“K“) and lambda (“l”), based on the amino acid sequences of their constant domains.
  • K“ kappa
  • l lambda
  • immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated alpha (“a“), delta (“5“), epsilon (““), gamma (“y“), and mu (“m”), respectively.
  • the g and a classes are further divided into subclasses (isotypes) on the basis of relatively minor differences in the CH sequence and function, e.g., humans express the following subclasses: IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2.
  • subclasses immunoglobulins
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known and described generally in, for example, Abbas et al. , Cellular and Molecular Immunology, 4 th ed. (W.B. Saunders Co., 2000).
  • The“variable region” or“variable domain” of an antibody refers to the amino-terminal domains of the heavy or light chain of the antibody.
  • the variable domains of the heavy chain and light chain may be referred to as“VH” and “VL”, respectively. These domains are generally the most variable parts of the antibody (relative to other antibodies of the same class) and contain the antigen binding sites.
  • variable refers to the fact that certain segments of the variable domains differ extensively in sequence among antibodies, such as anti-SIRP 1 antibodies of the present disclosure.
  • the variable domain mediates antigen binding and defines the specificity of a particular antibody for its particular antigen.
  • HVRs hypervariable regions
  • FR framework regions
  • the variable domains of native heavy and light chains each comprise four FR regions, largely adopting a beta-sheet configuration, connected by three HVRs, which form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • the HVRs in each chain are held together in close proximity by the FR regions and, with the HVRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Immunological Interest, Fifth Edition, National Institute of Health, Bethesda, MD (1991)).
  • the constant domains are not involved directly in the binding of antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody- dependent-cellular toxicity.
  • the term“monoclonal antibody” as used herein refers to an antibody, such as a monoclonal anti-SIRP 1 antibody of the present disclosure, obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations and/or post-translation modifications (e.g., isomerizations, amidations, etc.) that may be present in minor amounts.
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. In contrast to polyclonal antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • the modifier“monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including, for example, the hybridoma method, recombinant DNA methods, and technologies for producing human or human-like antibodies in animals that have parts or all of the human immunoglobulin loci or genes encoding human immunoglobulin sequences.
  • full-length antibody “intact antibody” or“whole antibody” are used interchangeably to refer to an antibody, such as an anti- SIRP 1 antibody of the present disclosure, in its substantially intact form, as opposed to an antibody fragment.
  • whole antibodies include those with heavy and light chains including an Fc region.
  • the constant domains may be native sequence constant domains (e.g ., human native sequence constant domains) or amino acid sequence variants thereof.
  • the intact antibody may have one or more effector functions.
  • an“antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include Fab, Fab', F(ab')2 and Fv fragments; diabodies; linear antibodies (see U.S. Patent 5641870, Example 2; Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called“Fab” fragments, and a residual“Fc” fragment, a designation reflecting the ability to crystallize readily.
  • the Fab fragment consists of an entire light chain along with the variable region domain of the heavy chain (VH), and the first constant domain of one heavy chain (Ciil).
  • VH variable region domain of the heavy chain
  • Ciil first constant domain of one heavy chain
  • Each Fab fragment is monovalent with respect to antigen binding, i.e., it has a single antigen-binding site.
  • Pepsin treatment of an antibody yields a single large F(ab') 2 fragment which roughly corresponds to two disulfide linked Fab fragments having different antigen-binding activity and is still capable of cross-linking antigen.
  • Fab' fragments differ from Fab fragments by having a few additional residues at the carboxy terminus of the Cn 1 domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the Fc fragment comprises the carboxy -terminal portions of both heavy chains held together by disulfides.
  • the effector functions of antibodies are determined by sequences in the Fc region, the region which is also recognized by Fc receptors (FcR) found on certain types of cells.
  • FcR Fc receptors
  • “Functional fragments” of antibodies, such as anti-SIRP 1 antibodies of the present disclosure comprise a portion of an intact antibody, generally including the antigen binding or variable region of the intact antibody or the Fc region of an antibody which retains or has modified FcR binding capability. Examples of antibody fragments include linear antibody, single-chain antibody molecules and multispecific antibodies formed from antibody fragments.
  • diabodies refers to small antibody fragments prepared by constructing sFv fragments (see preceding paragraph) with short linkers (about 5-10) residues) between the VH and VL domains such that inter-chain but not intra-chain pairing of the variable domains is achieved, thereby resulting in a bivalent fragment, i.e., a fragment having two antigen-binding sites.
  • Bispecific diabodies are heterodimers of two“crossover” sFv fragments in which the VH and VL domains of the two antibodies are present on different polypeptide chains.
  • a“chimeric antibody” refers to an antibody (immunoglobulin), such as a chimeric anti-SIRP 1 antibody of the present disclosure, in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • an antibody immunoglobulin
  • a chimeric anti-SIRP 1 antibody of the present disclosure in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is(are) identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody
  • Chimeric antibodies of interest herein include PRIMATIZED ® antibodies wherein the antigen-binding region of the antibody is derived from an antibody produced by, e.g., immunizing macaque monkeys with an antigen of interest.
  • “humanized antibody” is used a subset of“chimeric antibodies.”
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies comprising amino acid residues from non-human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • A“humanized form” of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • A“human antibody” is one that possesses an amino-acid sequence corresponding to that of an antibody, such as an anti-SIRP 1 antibody of the present disclosure, produced by a human and/or has been made using any of the techniques for making human antibodies as disclosed herein.
  • This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • Human antibodies can be produced using various techniques known in the art, including phage-display libraries and yeast-based platform technologies.
  • Human antibodies can be prepared by administering the antigen to a transgenic animal that has been modified to produce such antibodies in response to antigenic challenge, but whose endogenous loci have been disabled, e.g., immunized xenomice as well as generated via a human B-cell hybridoma technology.
  • hypervariable region refers to the regions of an antibody -variable domain, such as that of an anti-SIRP 1 antibody of the present disclosure, that are hypervariable in sequence and/or form structurally defined loops.
  • antibodies comprise six HVRs; three in the VH (Hl, H2, H3), and three in the VL (Ll, L2, L3).
  • H3 and L3 display the most diversity of the six HVRs, and H3 in particular is believed to play a unique role in conferring fine specificity to antibodies.
  • Naturally occurring camelid antibodies consisting of a heavy chain only are functional and stable in the absence of light chain.
  • the HVRs may be Rabat complementarity-determining regions (CDRs) based on sequence variability and are the most commonly used (Rabat et al., supra).
  • the HVRs may be Chothia CDRs. Chothia refers instead to the location of the structural loops (Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)).
  • the HVRs may be AbM HVRs. The AbM HVRs represent a compromise between the Rabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody-modeling software.
  • the HVRs may be“contact” HVRs. The“contact” HVRs are based on an analysis of the available complex crystal structures. The residues from each of these HVRs are noted below.
  • HVRs may comprise“extended HVRs” as follows: 24-36 or 24-34 (Ll), 46-56 or 50-56 (L2), and 89-97 or 89-96 (L3) in the VL, and 26-35 (Hl), 50-65 or 49-65 (a preferred embodiment) (H2), and 93-102, 94-102, or 95-102 (H3) in the VH.
  • the variable-domain residues are numbered according to Rabat el al., supra, for each of these extended-HVR definitions.
  • “Framework” or“FR” residues are those variable-domain residues other than the HVR residues as herein defined.
  • An“acceptor human framework” as used herein is a framework comprising the amino acid sequence of a VL or VH framework derived from a human immunoglobulin framework or a human consensus framework.
  • An acceptor human framework“derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may comprise pre-existing amino acid sequence changes. In some embodiments, the number of pre-existing amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • A“human consensus framework” is a framework that represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991). Examples include for the VL, the subgroup may be subgroup kappa I, kappa II, kappa III or kappa IV as in Kabat et al, supra. Additionally, for the VH, the subgroup may be subgroup I, subgroup II, or subgroup III as in Kabat et al, supra.
  • An“amino-acid modification” at a specified position refers to the substitution or deletion of the specified residue, or the insertion of at least one amino acid residue adjacent the specified residue. Insertion“adjacent” to a specified residue means insertion within one to two residues thereof. The insertion may be N-terminal or C- terminal to the specified residue.
  • the preferred amino acid modification herein is a substitution.
  • An“affinity-matured” antibody such as an affinity matured anti-SIRP 1 antibody of the present disclosure, is one with one or more alterations in one or more HVRs thereof that result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody that does not possess those alteration(s).
  • an affinity -matured antibody has nanomolar or even picomolar affinities for the target antigen.
  • Affinity-matured antibodies are produced by procedures known in the art. For example, Marks et al. Bio/Technology 10:779-783 (1992) describes affinity maturation by VH- and VL-domain shuffling.
  • Random mutagenesis of HVR and/or framework residues is described by, for example: Barbas et al. Proc Nat. Acad. Sci. USA 91 :3809-3813 (1994); Schier et al. Gene 169: 147-155 (1995); Yelton et al. J. Immunol. 155: 1994-2004 (1995); Jackson et al. J. Immunol. 154(7):3310-9 (1995); and Hawkins et al, J. Mol. Biol. 226:889-896 (1992).
  • Fv is the minimum antibody fragment which comprises a complete antigen-recognition and -binding site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervariable loops (3 loops each from the H and F chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three HVRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • Single-chain Fv also abbreviated as“sFv” or“scFv” are antibody fragments that comprise the VH and VL antibody domains connected into a single polypeptide chain.
  • the sFv polypeptide further comprises a polypeptide linker between the VH and VL domains, which enables the sFv to form the desired structure for antigen binding.
  • Antibody“effector functions” refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody, and vary with the antibody isotype.
  • the term“Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native-sequence Fc regions and variant Fc regions.
  • the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy-chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
  • the C-terminal lysine (residue 447 according to the EU numbering system) of the Fc region may be removed, for example, during production or purification of the antibody, or by recombinantly engineering the nucleic acid encoding a heavy chain of the antibody.
  • composition of intact antibodies may comprise antibody populations with all K447 residues removed, antibody populations with no K447 residues removed, and antibody populations having a mixture of antibodies with and without the K447 residue.
  • Suitable native-sequence Fc regions for use in the antibodies of the present disclosure include human IgGl, IgG2, IgG3 and IgG4.
  • A“native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • Native sequence human Fc regions include a native sequence human IgGl Fc region (non-A and A allotypes); native sequence human IgG2 Fc region; native sequence human IgG3 Fc region; and native sequence human IgG4 Fc region as well as naturally occurring variants thereof.
  • A“variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, preferably one or more amino acid substitution(s).
  • the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% homology with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% homology therewith, more preferably at least about 95% homology therewith.
  • Fc receptor or“FcR” describes a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and FcyRIII subclasses, including allelic variants and alternatively spliced forms of these receptors, FcyRI I receptors include FcyRIIA (an“activating receptor”) and FcyRIIB (an“inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor FcyRIIA contains an immunoreceptor tyrosine-based activation motif (“IT AM”) in its cytoplasmic domain.
  • Inhibiting receptor FcyRIIB contains an immunoreceptor tyrosine-based inhibition motif (“ITIM”) in its cytoplasmic domain.
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • Other FcRs including those to be identified in the future, are encompassed by the term“FcR” herein. FcRs can also increase the serum half-life of antibodies.
  • “percent (%) amino acid sequence identity” and“homology” with respect to a peptide, polypeptide or antibody sequence refers to the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific peptide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or MEGALIGNTM (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms known in the art needed to achieve maximal alignment over the full-length of the sequences being compared.
  • the term“compete” when used in the context of antibodies (e.g ., neutralizing antibodies) that compete for the same epitope means competition between antibody as determined by an assay in which the antibody being tested prevents or inhibits (e.g., reduces) specific binding of a reference molecule (e.g., a ligand, or a reference antibody) to a common antigen (e.g., SIRP 1 or a fragment thereof).
  • a reference molecule e.g., a ligand, or a reference antibody
  • a common antigen e.g., SIRP 1 or a fragment thereof.
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see, e.g., Stahli el al, 1983, Methods in Enzymology 9:242-253
  • solid phase direct biotin-avidin EIA see, e.g., Kirkland et al, 1986, J. Immunol.
  • solid phase direct labeled assay solid phase direct labeled sandwich assay (see, e.g., Harlow and Lane, 1988, Antibodies, A Laboratory Manual, Cold Spring Harbor Press); solid phase direct label RIA using 1-125 label (see, e.g., Morel et al, 1988, Molec. Immunol. 25:7-15); solid phase direct biotin-avidin EIA (see, e.g., Cheung, et al,
  • such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test antibody and a labeled reference antibody.
  • Antibodies identified by competition assay include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur. Additional details regarding methods for determining competitive binding are provided in the examples herein.
  • a competing antibody when present in excess, it will inhibit (e.g ., reduce) specific binding of a reference antibody to a common antigen by at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97.5%, and/or near 100%.
  • an“interaction” between a SIRP 1 polypeptide and a second polypeptide encompasses, without limitation, protein-protein interaction, a physical interaction, a chemical interaction, binding, covalent binding, and ionic binding.
  • the interaction can be inhibited by at least about any of 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97.5%, and/or near 100%.
  • epitope includes any determinant capable of being bound by an antibody.
  • An epitope is a region of an antigen that is bound by an antibody that targets that antigen, and when the antigen is a polypeptide, includes specific amino acids that directly contact the antibody. Most often, epitopes reside on polypeptides, but in some instances, can reside on other kinds of molecules, such as nucleic acids.
  • Epitope determinants can include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl or sulfonyl groups, and can have specific three- dimensional structural characteristics, and/or specific charge characteristics.
  • antibodies specific for a particular target antigen will preferentially recognize an epitope on the target antigen in a complex mixture of polypeptides and/or macromolecules.
  • An“agonist” antibody or an“activating” antibody is an antibody that induces (e.g., increases) one or more activities or functions of the antigen after the antibody binds the antigen.
  • An“antagonist” antibody or a“blocking” antibody or an“inhibitory” antibody is an antibody that reduces, inhibits, and/or eliminates (e.g., decreases) antigen binding to one or more ligand after the antibody binds the antigen, and/or that reduces, inhibits, and/or eliminates (e.g., decreases) one or more activities or functions of the antigen after the antibody binds the antigen.
  • antagonist antibodies, or blocking antibodies, or inhibitory antibodies substantially or completely inhibit antigen binding to one or more ligand and/or one or more activities or functions of the antigen.
  • An“isolated” antibody such as an isolated anti-SIRP 1 antibody of the present disclosure, is one that has been identified, separated and/or recovered from a component of its production environment (e.g., naturally or recombinantly).
  • the isolated antibody is free of association with all other contaminant components from its production environment.
  • Contaminant components from its production environment such as those resulting from recombinant transfected cells, are materials that would typically interfere with research, diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the antibody will be purified: (1) to greater than 95% by weight of antibody as determined by, for example, the Lowry method, and in some embodiments, to greater than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated antibody includes the antibody in situ within recombinant T-cells since at least one component of the antibody’s natural environment will not be present. Ordinarily, however, an isolated polypeptide or antibody will be prepared by at least one purification step.
  • An“isolated” nucleic acid molecule encoding an antibody is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the environment in which it was produced.
  • the isolated nucleic acid is free of association with all components associated with the production environment.
  • the isolated nucleic acid molecules encoding the polypeptides and antibodies herein is in a form other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from nucleic acid encoding the polypeptides and antibodies herein existing naturally in cells.
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • a“plasmid” refers to a circular double stranded DNA into which additional DNA segments may be ligated.
  • a phage vector refers to a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as“recombinant expression vectors,” or simply,“expression vectors.”
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • “plasmid” and“vector” may be used interchangeably as the plasmid is the most commonly used form of vector.
  • Polynucleotide or“nucleic acid,” as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase or by a synthetic reaction.
  • A“host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation.
  • a host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
  • Carriers as used herein include pharmaceutically acceptable carriers, excipients, or stabilizers that are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed.
  • the term“preventing” includes providing prophylaxis with respect to occurrence or recurrence of a particular disease, disorder, or condition in an individual.
  • An individual may be predisposed to, susceptible to a particular disease, disorder, or condition, or at risk of developing such a disease, disorder, or condition, but has not yet been diagnosed with the disease, disorder, or condition.
  • an individual“at risk” of developing a particular disease, disorder, or condition may or may not have detectable disease or symptoms of disease, and may or may not have displayed detectable disease or symptoms of disease prior to the treatment methods described herein.
  • At risk denotes that an individual has one or more risk factors, which are measurable parameters that correlate with development of a particular disease, disorder, or condition, as known in the art.
  • An individual having one or more of these risk factors has a higher probability of developing a particular disease, disorder, or condition than an individual without one or more of these risk factors.
  • treatment refers to clinical intervention designed to alter the natural course of the individual being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of progression, ameliorating or palliating the pathological state, and remission or improved prognosis of a particular disease, disorder, or condition.
  • An individual is successfully“treated”, for example, if one or more symptoms associated with a particular disease, disorder, or condition are mitigated or eliminated.
  • an“effective amount” refers to at least an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • An effective amount can be provided in one or more administrations.
  • An effective amount herein may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the treatment to elicit a desired response in the individual.
  • An effective amount is also one in which any toxic or detrimental effects of the treatment are outweighed by the therapeutically beneficial effects.
  • beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include clinical results such as decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival.
  • An effective amount of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective amount of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an “effective amount” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • An“individual” for purposes of treatment, prevention, or reduction of risk refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sport, or pet animals, such as dogs, horses, rabbits, cattle, pigs, hamsters, gerbils, mice, ferrets, rats, cats, and the like. In some embodiments, the individual is human.
  • administration“in conjunction” with another compound or composition includes simultaneous administration and/or administration at different times.
  • Administration in conjunction also encompasses administration as a co-formulation or administration as separate compositions, including at different dosing frequencies or intervals, and using the same route of administration or different routes of administration.
  • administration in conjunction is administration as a part of the same treatment regimen.
  • Antibodies provided are useful, e.g., for the diagnosis or treatment of the SIRP 1 mediated disorders.
  • an anti-SIRP 1 antibody of the present disclosure is an agonist of SIRP 1 activity.
  • an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1 but does not bind to human SIRPa.
  • an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1 but does not bind to human SIRPy.
  • an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1 but does not bind to human SIRP 1 isoform 3.
  • an anti-SIRP 1 antibody that binds to human SIRP 1 isoform 1 but does not bind to mouse SIRP 1 and/or cynomolgus monkey SIRP 1.
  • an anti-SIRP 1 antibody of the present disclosure agonizes SIRP 1 activity on CD 14-positive monocytes.
  • an anti- SIRP 1 antibody of the present disclosure induces respiratory burst in immune cells, such as neutrophils, monocytes, and/or macrophages.
  • an anti-SIRP 1 antibody of the present disclosure induces or increases IL-8 expression in monocytes.
  • an anti-SIRP 1 antibody of the present disclosure induces or increases TNFa expression in
  • an anti-SIRP 1 antibody of the present disclosure induces neutrophil-mediated phagocytosis, for example, of tumor cells. In some embodiments, an anti-SIRP 1 antibody of the present disclosure increases neutrophil-mediated tumor cell clearance. In some embodiments, an anti-SIRP 1 antibody of the present disclosure increases or enhances anti-tumor properties or activity of neutrophils.
  • an anti-SIRP 1 antibody of the present disclosure recruits immune cells. In some embodiments, an anti-SIRP 1 antibody of the present disclosure induces syk phosphorylation. In some embodiments, an anti-SIRP 1 antibody of the present disclosure induces syk phosphorylation when clustered by adjacent cells expressing Fc gamma receptors.
  • SIRP 1 antibody of the present disclosure downregulates SIRP 1 expression on the surface of a cell. In some embodiments, SIRP 1 antibody of the present disclosure does not downregulate SIRP 1 expression on the surface of a cell. In some embodiments, SIRP l antibody of the present disclosure blocks binding of a ligand to SIRP l. In some embodiments, SIRP 1 antibody of the present disclosure does not block binding of a ligand to SIRP L
  • an anti-SIRP 1 antibody is provided that has one or more properties selected from:
  • g induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro and/or in vivo,
  • neutrophil-mediated phagocytosis for example, of tumor cells in vitro and/or in vivo
  • m increases viability of macrophages in vitro and/or in vivo, alone and/or in combination with an agonist anti-TREM2 antibody
  • n increases viability of dendritic cells in vitro and/or in vivo.
  • an anti-SIRP 1 antibody is provided that has the following properties:
  • g induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro and/or in vivo,
  • neutrophil-mediated phagocytosis for example, of tumor cells in vitro and/or in vivo
  • anti-SIRP 1 antibodies comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (b) HVR-H2 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (c) HVR-H3 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (d) HVR-L1 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8; (e) HVR-L2 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8; and (f) HVR-L3 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8.
  • anti-SIRP 1 antibodies comprising HVR-H1, HVR-H2, HVR-H3, HVR-L1, HVR-L2, and HVR-L3, wherein: (a) HVR-H1 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (b) HVR-H2 comprising an amino acid sequence of an HVR-H2 shown in Table 4 and/or Table 9; (c) HVR-H3 comprising an amino acid sequence of an HVR-H3 shown in Table 4 and/or Table 9; (d) HVR-L1 comprising an amino acid sequence of an HVR-L1 shown in Table 3 and/or Table 8; (e) HVR-L2 comprising an amino acid sequence of an HVR-L2 shown in Table 3 and/or Table 8; and (f) HVR-L3 comprising an amino acid sequence of an HVR-L3 shown in Table 3 and/or Table 8.
  • anti-SIRP 1 antibodies comprising six HVRs of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB-17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-24,
  • SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3, 4, 8, and 9).
  • anti-SIRP 1 antibodies comprising six HVRs of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3, 4, 8, and 9).
  • anti-SIRP 1 antibodies comprising six HVRs of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB- 2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 8 and 9).
  • anti-SIRP 1 antibodies comprising six HVRs of an antibody selected from SB-l-3, SB-2-8, and SB-8-13 (shown in Tables 8 and 9).
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 228; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 238; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 239; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 240; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 241; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 230; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 242; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 231; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 243; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 232; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 244; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 99; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 245; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 230; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 242; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383;
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16;
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 246; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 234; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 247; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 248; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 246; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383;
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19;
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 235; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 249; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 2
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 22
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 236; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 250; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 2
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 22
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 237; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 251; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 2
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 22
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • anti-SIRP 1 antibodies comprising (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 236; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 252; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO:
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 2
  • HVR-L2 comprising the amino acid sequence of SEQ ID NO: 22
  • HVR-L3 comprising the amino acid sequence of SEQ ID NO: 69.
  • anti-SIRP 1 antibodies comprising at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (b) HVR-H2 comprising an amino acid sequence of an HVR-H2 shown in Table 4 and/or Table 9; (c) HVR-H3 comprising an amino acid sequence of an HVR-H3 shown in Table 4 and/or Table 9.
  • an anti-SIRP 1 antibody comprises HVR-H1, HVR, H2, and HVR-H3 of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB-17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-24, SB-25, SB-26, SB-27, SB-28, SB-29, SB-30, SB-31, SB-32, SB-33, SB-34, SB-35, SB-36, SB-37, SB-38, SB-39, SB-40, SB-41, SB-42, SB-43, SB-44, SB-45, SB-46, SB-47, SB-48, SB-49, SB-50, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2- 7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13
  • an anti-SIRPpl antibody comprises HVR-H1, HVR, H2, and HVR-H3 of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 4 and 9).
  • an anti-SIRP 1 antibody comprises HVR-H1, HVR, H2, and HVR-H3 of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Table 9).
  • an anti-SIRP 1 antibody comprises HVR-H1, HVR, H2, and HVR-H3 of an antibody selected from SB-l-3, SB-2-8, and SB-8- 13 (shown in Table 9).
  • an anti- SIRP 1 antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 239; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 125.
  • an anti-SIRP 1 antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 231; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 243; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 126.
  • an anti-SIRP 1 antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 246; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 131.
  • anti-SIRP 1 antibodies comprising at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising an amino acid sequence of an HVR-L1 shown in Table 3 and/or Table 8; (b) HVR-L2 comprising an amino acid sequence of an HVR-L2 shown in Table 3 and/or Table 8; (c) HVR-L3 comprising an amino acid sequence of an HVR-L3 shown in Table 3 and/or Table 8.
  • an anti-SIRP 1 antibody comprises HVR-L1, HVR, L2, and HVR-L3 of an antibody selected from SB-l, SB-2, SB- 3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB- 17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-24, SB-25, SB-26, SB-27, SB-28, SB-29, SB- 30, SB-31, SB-32, SB-33, SB-34, SB-35, SB-36, SB-37, SB-38, SB-39, SB-40, SB-41, SB-42, SB- 43, SB-44, SB-45, SB-46, SB-47, SB-48, SB-49, SB-50, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13
  • an anti-SIRP l antibody comprises HVR-L1, HVR, L2, and HVR-L3 of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l -2, SB-l-3, SB-l-4, SB- 1-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3 and 8).
  • an anti- SIRP 1 antibody comprises HVR-L1, HVR, L2, and HVR-L3 of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Table 8).
  • an anti-SIRP 1 antibody comprises HVR-L1, HVR, L2, and HVR-L3 of an antibody selected from SB-l-3, SB-2-8, and SB-8-13 (shown in Table 8).
  • an anti-SIRP l antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • an anti-SIRP 1 antibody comprises (a) HVR- Ll comprising the amino acid sequence of SEQ ID NO: 383; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • an anti-SIRP 1 antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • anti-SIRP 1 antibodies comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- Hl comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9, (ii) HVR- H2 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9, and (iii) HVR-H3 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8, (ii) HVR-L2 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8, and (iii) HVR-L3 comprising an amino acid sequence of
  • anti-SIRP 1 antibodies comprising (a) a VH domain comprising (i) HVR-H1 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9, (ii) HVR-H2 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9, and (iii) HVR-H3 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; and (b) a VL domain comprising (i) HVR-L1 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8, (ii) HVR-L2 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8, and (iii) HVR-L3 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8.
  • anti-SIRP 1 antibodies comprising a VH domain comprising HVR-H1, HVR-H2, and HVR-H2, and a VL domain comprising HVR-L1, HVR-L2, and HVR-L3, wherein HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 are the HVRs of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l,
  • SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3, 4, 8, and 9).
  • anti-SIRP 1 antibodies comprising a VH domain comprising HVR-H1, HVR-H2, and HVR-H2, and a VL domain comprising HVR-L1, HVR-L2, and HVR-L3, wherein HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 are the HVRs of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 3, 4, 8, and 9).
  • anti-SIRP 1 antibodies comprising a VH domain comprising HVR-H1, HVR-H2, and HVR-H2, and a VL domain comprising HVR-L1, HVR-L2, and HVR-L3, wherein HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 are the HVRs of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB- 2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21 (shown in Tables 8 and 9).
  • anti-SIRP 1 antibodies comprising a VH domain comprising HVR-H1, HVR-H2, and HVR-H2, and a VL domain comprising HVR-L1, HVR-L2, and HVR-L3, wherein HVR-H1, HVR-H2, HVR-H2, HVR-L1, HVR-L2, and HVR-L3 are the HVRs of an antibody selected from SB-l-3, SB-2-8, and SB-8-13 (shown in Tables 8 and 9).
  • anti-SIRP 1 antibodies comprising (a) a VH domain comprising (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 229; (ii) ETVR- H2 comprising the amino acid sequence of SEQ ID NO: 239; (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 125; and (b) a VL domain comprising (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 31.
  • anti-SIRP 1 antibodies comprising (a) a VH domain comprising (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 231; (ii) ETVR- H2 comprising the amino acid sequence of SEQ ID NO: 243; (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 126; and (b) a VL domain comprising (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 16; and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 32.
  • anti-SIRP 1 antibodies comprising (a) a VH domain comprising (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 233; (ii) ETVR- H2 comprising the amino acid sequence of SEQ ID NO: 246; (iii) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 131; and (b) a VL domain comprising (iv) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 383; (v) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 19; and (vi) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 37.
  • an anti-SIRP 1 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 268, 270, 272, 274, 276, 279, 281,
  • VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321,
  • 363, or 365 contains substitutions (e.g ., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-SIRP 1 antibody comprising that sequence retains the ability to bind to SIRP 1.
  • a total of 1 to 10 amino acids have been substituted, inserted, and/or deleted in SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325, 327, 329, 331,
  • a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 268, 270, 272,
  • the anti-SIRP 1 antibody comprises the VH sequence of SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325, 327, 329, 331, 333, 335, 337, 339, 341, 343, 345, 347, 349, 351, 353, 355, 357, 359, 361, 363, 365, 366, 367, 368, 369, 371, 372, 373, 374,
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (b) HVR-H2 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9; (c) HVR-H3 comprising an amino acid sequence of an HVR-H1 shown in Table 4 and/or Table 9.
  • an anti-SIRP 1 antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 267, 269, 271, 273,
  • VL light chain variable domain
  • VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 267, 269, 271, 273, 275, 277, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302,
  • 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, or 370 contains substitutions ( e.g ., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti- SIRP 1 antibody comprising that sequence retains the ability to bind to SIRP 1.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO:
  • the anti- SIRP 1 antibody comprises the VL sequence of SEQ ID NO: 267, 269, 271, 273, 275, 277, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302, 304, 306, 308, 310, 312, 314, 316, 318, 320, 322, 324, 326, 328, 330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, or 370.
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti- SIRP 1 antibody comprises the VL sequence of SEQ ID NO: 267, 269, 271, 273, 275, 277, 278, 280, 282, 284, 286, 288, 290, 292, 294, 296, 298, 300, 302, 304, 306, 308, 310, 312,
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8; (b) HVR-L2 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8; and (c) HVR-L3 comprising an amino acid sequence of an HVR-H1 shown in Table 3 and/or Table 8.
  • the anti-SIRP 1 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the
  • anti-SIRP 1 antibodies wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises a VH sequence selected from SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325, 327, 329, 331, 333, 335,
  • the antibody comprises the VH sequence and VL sequence of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB-17, SB-18, SB-19, SB-20, SB-21,
  • the antibody comprises the VH sequence and VL sequence of an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49,
  • the antibody comprises the VH sequence and VL sequence of an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody comprises the VH sequence and VL sequence of an antibody selected from SB-l-2, SB-l-3,
  • the antibody comprises the VH sequence and VL sequence of an antibody selected from SB-l-3, SB-2-8, and SB-8-13. In some embodiments, the antibody comprises the VH sequence of SEQ ID NO: 367 and VL sequence of SEQ ID NO: 267. In some embodiments, the antibody comprises the VH sequence of SEQ ID NO: 372 and VL sequence of SEQ ID NO: 370. In some embodiments, the antibody comprises the VH sequence of SEQ ID NO: 376 and VL sequence of SEQ ID NO: 280.
  • an anti-SIRP 1 antibody competes for binding with an antibody comprising a VH sequence selected from SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325,
  • an anti-SIRP 1 antibody competes for binding with an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB-l l, SB-12, SB-13, SB-14, SB-15, SB-16, SB- 17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-24, SB-25, SB-26, SB-27,
  • an anti-SIRP l antibody competes for binding with an antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9,
  • SB-2-10 SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-
  • an anti-SIRP 1 antibody competes for binding with an antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8- 13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • an anti-SIRP 1 antibody competes for binding with an antibody selected from SB-l- 3, SB-2-8, and SB-8-13.
  • an anti-SIRP 1 antibody competes for binding with an antibody comprising the VH sequence of SEQ ID NO: 367 and VL sequence of SEQ ID NO: 267.
  • an anti-SIRP 1 antibody competes for binding with an antibody comprising the VH sequence of SEQ ID NO: 372 and VL sequence of SEQ ID NO: 370. In some embodiments, an anti-SIRP 1 antibody competes for binding with an antibody comprising the VH sequence of SEQ ID NO: 376 and VL sequence of SEQ ID NO: 280.
  • the antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody comprising a VH sequence selected from SEQ ID NO: 268, 270, 272, 274, 276, 279, 281, 283, 285, 287, 289, 291, 293, 295, 297, 299, 301, 303, 305, 307, 309, 311, 313, 315, 317, 319, 321, 323, 325, 327, 329, 331, 333, 335, 337, 339,
  • VL sequence selected from SEQ ID NO: 267,
  • the antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti- SIRP l antibody selected from SB-l, SB-2, SB-3, SB-4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-10, SB- 11, SB-12, SB-13, SB-14, SB-15, SB-16, SB-17, SB-18, SB-19, SB-20, SB-21, SB-22, SB-23, SB-
  • SB-8-15 SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody selected from SB-l, SB-2, SB-3, SB- 4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8-13, SB-8-14, SB-8-15, SB-8-16, SB-40- 18, SB-40- 19, SB-40-20, and SB-40-21.
  • an anti-SIRP 1 antibody selected from SB-l, SB-2, SB-3, SB- 4, SB-5, SB-6, SB-7, SB-8, SB-9, SB-14, SB-28, SB-39, SB-40, SB-49, SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB
  • the antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody selected from SB-l-2, SB-l-3, SB-l-4, SB-l-5, SB-2-7, SB-2-8, SB-2-9, SB-2-10, SB-2-11, SB-8- 13, SB-8-14, SB-8-15, SB-8-16, SB-40-18, SB-40-19, SB-40-20, and SB-40-21.
  • the antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP l antibody selected from SB-l-3, SB-2-8, and SB-8-13.
  • an anti-SIRP 1 antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody comprising the VH sequence of SEQ ID NO: 367 and VL sequence of SEQ ID NO: 267. In some embodiments, an anti-SIRP 1 antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody comprising the VH sequence of SEQ ID NO: 372 and VL sequence of SEQ ID NO: 370.
  • an anti-SIRP 1 antibody binds to an epitope of human SIRP 1 that is the same as or overlaps with the epitope bound by an anti-SIRP 1 antibody comprising the VH sequence of SEQ ID NO: 376 and VL sequence of SEQ ID NO: 280.
  • the epitope of human SIRP 1 is the same epitope as bound by an anti-SIRP 1 antibody.
  • an anti-SIRP 1 antibody binds to an epitope within amino acids 30 to 148 of SEQ ID NO: 1. In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 30 to 136 of SEQ ID NO: 1. In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 30 to 80 of SEQ ID NO: 1. In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 40 to 90 of SEQ ID NO: 1. In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 50 to 100 of SEQ ID NO: 1.
  • an anti-SIRP 1 antibody binds to an epitope within amino acids 60 to 110 of SEQ ID NO: l._ In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 70 to 120 of SEQ ID NO: l._ In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 80 to 130 of SEQ ID NO: l._ In some embodiments, an anti-SIRP 1 antibody binds to an epitope within amino acids 90 to 140 of SEQ ID NO: 1. _
  • the anti-SIRP 1 antibody according to any of the above embodiments is a monoclonal antibody, including a humanized and/or human antibody.
  • the anti- SIRP 1 antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab')2 fragment.
  • the anti-SIRP 1 antibody is a substantially full-length antibody, e.g., an IgGl antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • an anti-SIRP 1 antibody may incorporate any of the features, singly or in combination, as described in Sections 1-7 below: (1) Anti-SIRPf ) 1 antibody binding affinity
  • the antibody has a dissociation constant (KD) of ⁇ 1 mM, ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM ( e.g ., 10 8 M or less, e.g., from 10 8 M to 10 13 M, e.g., from 10 9 M to 10 13 M).
  • KD dissociation constant
  • Dissociation constants may be determined through any analytical technique, including any biochemical or biophysical technique such as ELISA, surface plasmon resonance (SPR), bio-layer interferometry (see, e.g., Octet System by ForteBio), isothermal titration calorimetry (ITC), differential scanning calorimetry (DSC), circular dichroism (CD), stopped-flow analysis, and colorimetric or fluorescent protein melting analyses.
  • KD is measured by a radiolabeled antigen binding assay (RIA).
  • RIA radiolabeled antigen binding assay
  • an RIA is performed with the Fab version of an antibody of interest and its antigen, for example as described in Chen et al. J. Mol. Biol. 293:865-881(1999)).
  • KD is measured using a BIACORE surface plasmon resonance assay, for example, an assay using a BIACORE -2000 or a BIACORE -3000 (BIAcore, Inc., Piscataway, NJ) is performed at 25°C with immobilized antigen CM5 chips at -10 response units (RU).
  • KD is measured using a ForteBio Octet ® Red384 system (ForteBio, Menlo Park, CA), for example, as discussed in the examples herein.
  • the KD is determined using a monovalent antibody (e.g., a Fab) or a full-length antibody. In some embodiments, the KD is determined using a full-length antibody in a monovalent form.
  • a monovalent antibody e.g., a Fab
  • a full-length antibody in a monovalent form.
  • the antibody antibodies is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment antigen
  • Fab' fragment antigen binding domain 2
  • Fv fragment antigen binding domain 2
  • Fv fragment antigen binding domain 2
  • scFv fragments fragment antigen binding domains
  • other fragments described below For a review of certain antibody fragments, see Hudson et al. Nat. Med. 9: 129-134 (2003).
  • scFv fragments see. e.g., WO 93/16185; and U.S. Patent Nos. 5571894 and 5587458.
  • Fab and F(ab') 2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half- life, see U.S. Patent No. 5869046.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP404097; WO 1993/01161; Hudson et al. Nat. Med. 9: 129-134 (2003). Triabodies and tetrabodies are also described in Hudson et al. Nat. Med. 9: 129-134 (2003).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (see, e.g., U.S. Patent No. 6248516).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g., E . coli or phage), as described herein.
  • the antibody is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4816567.
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a“class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen binding fragments thereof.
  • the antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody is substantially non-immunogenic in humans.
  • a humanized antibody has substantially the same affinity for a target as an antibody from another species from which the humanized antibody is derived. See, e.g., U.S. Pat. No.
  • a humanized antibody comprises one or more variable domains in which HVRs (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), for example, to restore or improve antibody specificity or affinity.
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the“best- fit” method (see, e.g., Sims et al. J. Immunol. 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA 89:4285 (1992); and Presta et al., J. Immunol.
  • the antibody is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk et al. Curr. Opin. Pharmacol. 5:368-74 (2001) and Lonberg Curr. Opin. Immunol. 20:450-459 (2008).
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge.
  • Large human Ig fragments can preserve the large variable gene diversity as well as the proper regulation of antibody production and expression.
  • the reproduced human antibody repertoire in these mouse strains can yield high affinity fully human antibodies against any antigen of interest, including human antigens.
  • antigen-specific human MAbs with the desired specificity can be produced and selected.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol. 133:3001 (1984) and Boemer et al. J. Immunol. 147:86 (1991)). Human antibodies generated via human B-cell hybridoma technology are also described in Li et al. Proc. Natl. Acad. Sci. USA, 1 03:3557-3562 (2006). Additional methods include those described, for example, in U.S. Patent No. 7189826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines).
  • Human hybridoma technology (Trioma technology) is also described in Vollmers et al. Histology and Hisiopaihology 20(3) :927- 937 (2005) and Vollmers et al. Methods and Findings in Experimental and Clinical Pharmacology 27(3): 185-91 (2005).
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • the antibody is a human antibody isolated by in vitro methods and/or screening combinatorial libraries for antibodies with the desired activity or activities. Suitable examples include but are not limited to phage display (CAT, Morphosys, Dyax, Biosite/Medarex, Xoma, Symphogen, Alexion (formerly Proliferon), Affimed) ribosome display (CAT), yeast-based platform technology (Adimab), and the like.
  • repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen binding phage as described in Winter et al. Ann. Rev. Immunol. 12: 433-455 (1994).
  • PCR polymerase chain reaction
  • a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. See also Sidhu et al. J. Mol. Biol. 338(2): 299-310, 2004; Lee et al. J. Mol. Biol. 340(5): 1073-1093, 2004; Fellouse Proc. Natl. Acad. Sci.
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas.
  • the naive repertoire can be cloned ( e.g ., from human) to provide a single source of antibodies to a wide range of non-self and also self-antigens without any immunization as described by Griffiths et al. EMBO J. 12: 725- 734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers comprising random sequence to encode the highly variable FTVR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom et al. J. Mol. Biol., 227: 381-388, 1992.
  • Patent publications describing human antibody phage libraries include, for example: US Patent No. 5750373, and US Patent Publication Nos. 2007/0292936 and 2009/0002360.
  • Antibodies isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • the antibody comprises an Fc.
  • the Fc is a human IgGl, IgG2, IgG3, and/or IgG4 isotype.
  • the antibody is of the IgG class, the IgM class, or the IgA class.
  • the antibody has an IgG2 isotype. In some embodiments, the antibody contains a human IgG2 constant region. In some embodiments, the human IgG2 constant region includes an Fc region. In some embodiments, the antibody induces the one or more SIRP 1 activities or independently of binding to an Fc receptor. In some embodiments, the antibody binds an inhibitory Fc receptor. In certain embodiments, the inhibitory Fc receptor is inhibitory Fc-gamma receptor IIB (FcyllB). [0103] In certain embodiments of any of the antibodies provided herein, the antibody has an IgGl isotype. In some embodiments, the antibody contains a mouse IgGl constant region.
  • the antibody contains a human IgGl constant region.
  • the human IgGl constant region includes an Fc region.
  • the antibody binds an inhibitory Fc receptor.
  • the inhibitory Fc receptor is inhibitory Fc-gamma receptor IIB (FcyllB).
  • the antibody has an IgG4 isotype.
  • the antibody contains a human IgG4 constant region.
  • the human IgG4 constant region includes an Fc region.
  • the antibody binds an inhibitory Fc receptor.
  • the inhibitory Fc receptor is inhibitory Fc-gamma receptor IIB (FcyllB).
  • the antibody has a hybrid IgG2/4 isotype.
  • the antibody includes an amino acid sequence comprising amino acids 118 to 260 according to EU numbering of human IgG2 and amino acids 261-447 according to EU numbering of human IgG4 (WO 1997/11971; WO 2007/106585).
  • the Fc region increases clustering without activating complement as compared to a corresponding antibody comprising an Fc region that does not comprise the amino acid substitutions.
  • the antibody induces one or more activities of a target specifically bound by the antibody.
  • the antibody binds to SIRP 1.
  • an anti-SIRP 1 antibody of the present disclosure may also be desirable to modify effector function and/or to increase serum half-life of the antibody.
  • the Fc receptor binding site on the constant region may be modified or mutated to remove or reduce binding affinity to certain Fc receptors, such as FcyRI, FcyRII, and/or FcyRIII to reduce Antibody-dependent cell-mediated cytotoxicity.
  • the effector function is impaired by removing N- glycosylation of the Fc region (e.g ., in the CH2 domain of IgG) of the antibody.
  • the effector function is impaired by modifying regions such as 233-236, 297, and/or 327-331 of human IgG as described in WO 99/58572 and Armour et al. Molecular Immunology 40: 585-593 (2003); Reddy et al. J. Immunology 164: 1925-1933 (2000).
  • a salvage receptor binding epitope into the antibody (especially an antibody fragment) as described in U.S. Patent 5739277, for example.
  • the term“salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule ( e.g ., IgGi, IgG2, IgG3, or IgGi) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • IgGi an epitope of the Fc region of an IgG molecule
  • Multispecific are antibodies that have binding specificities for at least two different epitopes, including those on the same or another polypeptide (e.g., one or more SIRP 1 polypeptides of the present disclosure).
  • the multispecific antibody is a bispecific antibody.
  • the multispecific antibody is a trispecific antibody.
  • the multispecific antibody is a tetraspecific antibody.
  • Such antibodies can be derived from full-length antibodies or antibody fragments (e.g., F(ab’)2bispecific antibodies).
  • the multispecific antibody comprises a first antigen binding region which binds to first site on SIRP 1 and comprises a second antigen binding region which binds to a second site on SIRP 1.
  • the multispecific antibodies comprises a first antigen binding region which binds to SIRP 1 and a second antigen binding region that binds to a second polypeptide.
  • multispecific antibodies comprises a first antigen binding region, wherein the first antigen binding region comprises the six FTVRs of an antibody described herein, which binds to SIRP 1 and a second antigen binding region that binds to a second polypeptide.
  • the first antigen binding region comprises the VH or VL of an antibody described herein.
  • the second polypeptide is an antigen facilitating transport across the blood-brain-barrier.
  • antibody herein is conjugated to a peptide that facilitates transport across the blood-brain barrier.
  • Numerous antigens and peptides are known in the art that facilitate transport across the blood-brain barrier (see, e.g., Gabathuler R. Neurobiol. Dis. 37:48-57 (2010)).
  • Such second antigens and peptides include, without limitation, transferrin receptor (TR), insulin receptor (FUR), insulin-like growth factor receptor (IGFR), low-density lipoprotein receptor related proteins 1 and 2 (LPR-l and 2), diphtheria toxin receptor, including CRM197 (a non-toxic mutant of diphtheria toxin), TMEM 30(A)
  • polypeptide is transferrin.
  • the second polypeptide is a disease-causing protein selected from amyloid beta, oligomeric amyloid beta, amyloid beta plaques, amyloid precursor protein or fragments thereof, Tau, IAPP, alpha-synuclein, TDP-43, FUS protein, C9orf72 (chromosome 9 open reading frame 72), c9RAN protein, prion protein, PrPSc, huntingtin, calcitonin, superoxide dismutase, ataxin, ataxin 1, ataxin 2, ataxin 3, ataxin 7, ataxin 8, ataxin 10, Lewy body, atrial natriuretic factor, islet amyloid polypeptide, insulin, apolipoprotein AI, serum amyloid A, medin, prolactin, transthyretin, lysozyme, beta 2 microglobulin, gelsolin, keratoepithelin, cystatin, immunoglobulin light chain AL,
  • the second polypeptide is a ligand and/or protein expressed on immune cells, wherein the ligand and/or protein is selected from CD40, 0X40, ICOS, CD28, CD137/4-1BB, CD27, GITR, PD-L1, CTLA-4, PD-L2, PD-l, B7-H3, B7-H4, HVEM, BTLA, KIR, GAIN, TIM3, A2AR, LAG-3, and phosphatidylserme.
  • the ligand and/or protein is selected from CD40, 0X40, ICOS, CD28, CD137/4-1BB, CD27, GITR, PD-L1, CTLA-4, PD-L2, PD-l, B7-H3, B7-H4, HVEM, BTLA, KIR, GAIN, TIM3, A2AR, LAG-3, and phosphatidylserme.
  • the second polypeptide is a protein, lipid, polysaccharide, or glycolipid expressed on one or more tumor cells and any combination thereof.
  • the second polypeptide is an immunoglobulin-like receptor, such as TREM2. In some embodiments of any of the multispecific antibodies, the second polypeptide is an immunoglobulin-like receptor expressed on myeloid lineage cells.
  • the multivalent antibody contains at least one polypeptide chain (and preferably two polypeptide chains), wherein the polypeptide chain or chains comprise two or more variable domains.
  • the polypeptide chain or chains may comprise VDl-(Xl) n -VD2-(X2) n -Fc, wherein VD1 is a first variable domain, VD2 is a second variable domain, Fc is one polypeptide chain of an Fc region, XI and X2 represent an amino acid or polypeptide, and n is 0 or 1.
  • the polypeptide chain or chains may comprise VH-CH! -flexible linker- VH-CH!
  • the multivalent antibody herein preferably further comprises at least two (and preferably four) light chain variable domain polypeptides.
  • the multivalent antibody herein may, for instance, comprise from about two to about eight light chain variable domain polypeptides.
  • the light chain variable domain polypeptides contemplated here comprise a light chain variable domain and, optionally, further comprise a CL domain.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain- light chain pairs having different specificities (see Milstein and Cuello Nature 305: 537 (1983), WO 93/08829, and Traunecker et al. EMBO J. 10:3655 (1991)), and“knob-in-hole” engineering (see, e.g., U.S. Patent No. 5731168). See also WO 2013/026833 (CrossMab).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc- heterodimeric molecules (WO 2009/089004A1); cross- linking two or more antibodies (see, e.g., US Patent No. 4676980); using leucine; using“diabody” technology for making bispecific antibody fragments (see, e.g., Hollinger el al. Proc. Natl. Acad.
  • Engineered antibodies with three or more functional antigen binding sites are also included herein (see, e.g., US 2006/0025576).
  • the antibody herein also includes a“Dual Acting FAb” or“DAF” comprising an antigen binding site that binds to multiple SIRPpl (see, US 2008/0069820, for example).
  • amino acid sequence variants of the antibodies are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • antibody variants having one or more amino acid substitutions are provided.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. TABLE 1: Amino Acid Substitutions
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • non-conservative substitutions can involve the exchange of a member of one of these classes for a member from another class.
  • Such substituted residues can be introduced, for example, into regions of a human antibody that are homologous with non-human antibodies, or into the non-homologous regions of the molecule.
  • the hydropathic index of amino acids can be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. They are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5);
  • the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biologically functional protein or peptide thereby created is intended for use in immunological embodiments, as in the present case.
  • the greatest local average hydrophilicity of a protein as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein.
  • the substitution of amino acids whose hydrophilicity values are within ⁇ 2 is included, in certain embodiments, those which are within ⁇ 1 are included, and in certain embodiments, those within ⁇ 0.5 are included.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may, for example, be outside of antigen contacting residues in the HVRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • Amino acid sequence insertions include amino- and/or carboxyl -terminal fusions ranging in length from one residue to polypeptides comprising a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g., for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • cysteine residues not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment, such as an Fv fragment).
  • the antibody is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • X is any amino acid except proline
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 according to Kabat numbering of the CH2 domain of the Fc region.
  • the oligosaccharide may include various carbohydrates, for example, mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the“stem” of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. See, e.g., US Patent Publication Nos. 2003/0157108 and 2004/0093621.
  • Examples of publications related to“defucosylated” or“fucose- deficient” antibody variants include: US 2003/0157108; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865;
  • Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US 2003/0157108), and knockout cell lines, such as alpha- l,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004) and Kanda el al.
  • the antibody Fc is an antibody Fc isotype and/or modification. In some embodiments, the antibody Fc isotype and/or modification is capable of binding to Fc gamma receptor.
  • the modified antibody Fc is an IgGl modified Fc.
  • the IgGl modified Fc comprises one or more modifications.
  • the IgGl modified Fc comprises one or more amino acid substitutions (e.g relative to a wild-type Fc region of the same isotype).
  • the one or more amino acid substitutions are selected from N297A (Bolt S et al.
  • the Fc comprises N297A mutation according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises D265A and N297A mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises D270A mutations according to EU numbering. In some embodiments, the IgGl modified Fc comprises L234A and L235A mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises L234A and G237A mutations according to EU numbering.
  • the Fc comprises L234A, L235A and G237A mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises one or more (including all) of P238D, L328E, E233,
  • the Fc comprises one or more of S267E/L328F mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises P238D, L328E, E233D, G237D, H268D, P271G and A330R mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises P238D, L328E, G237D, H268D, P271G and A33 OR mutations according to EU numbering.
  • the Fc comprises P238D, S267E, L328E, E233D, G237D, H268D, P271G and A330R mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises P238D, S267E, L328E, G237D, H268D, P271G and A330R mutations according to EU numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises C226S, C229S, E233P, L234V, and L235A mutations according to EU numbering.
  • the Fc comprises L234F, L235E, and P331S mutations according to EFT numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises S267E and F328F mutations according to EFT numbering. In some embodiments of any of the IgGl modified Fc, the Fc comprises S267E mutations according to EFT numbering.
  • the Fc comprises a substitute of the constant heavy 1 (CH1) and hinge region of IgGl with CH1 and hinge region of IgG2 (amino acids 118-230 of IgG2 according to EFT numbering) with a Kappa light chain.
  • CH1 constant heavy 1
  • IgG2 amino acids 118-230 of IgG2 according to EFT numbering
  • the Fc includes two or more amino acid substitutions that increase antibody clustering without activating complement as compared to a corresponding antibody having an Fc region that does not include the two or more amino acid substitutions.
  • the IgGl modified Fc is an antibody comprising an Fc region, where the antibody comprises an amino acid substitution at position E430G and one or more amino acid substitutions in the Fc region at a residue position selected from: F234F, F235A, F235E, S267E, K322A, F328F, A330S, P331S, and any combination thereof according to EFT numbering.
  • the IgGl modified Fc comprises an amino acid substitution at positions E430G, F243A, F235A, and P331S according to EFT numbering. In some embodiments, the IgGl modified Fc comprises an amino acid substitution at positions E430G and P331 S according to EFT numbering. In some embodiments, the IgGl modified Fc comprises an amino acid substitution at positions E430G and K322A according to EFT numbering. In some embodiments, the IgGl modified Fc comprises an amino acid substitution at positions E430G, A330S, and P331S according to EFT numbering.
  • the IgGl modified Fc comprises an amino acid substitution at positions E430G, K322A, A330S, and P331S according to EFT numbering. In some embodiments, the IgGl modified Fc comprises an amino acid substitution at positions E430G, K322A, and A330S according to EFT numbering. In some embodiments, the IgGl modified Fc comprises an amino acid substitution at positions E430G, K322A, and P331S according to EFT numbering.
  • the IgGl modified Fc may further comprise herein may be combined with an A330F mutation (Fazar el al. Proc Natl Acad Sci USA, 103:4005-4010 (2006)), or one or more of F234F, F235E, and/or P331S mutations (Sazinsky et al. Proc Natl Acad Sci USA, 105:20167-20172 (2008)), according to the EU numbering convention, to eliminate complement activation.
  • A330F mutation Fazar el al. Proc Natl Acad Sci USA, 103:4005-4010 (2006)
  • F234F, F235E, and/or P331S mutations Sazinsky et al. Proc Natl Acad Sci USA, 105:20167-20172 (2008)
  • the IgGl modified Fc may further comprise one or more of A330F, A330S, F234F, F235E, and/or P331S according to EU numbering. In some embodiments of any of the IgGl modified Fc, the IgGl modified Fc may further comprise one or more mutations to enhance the antibody half-life in human serum (e.g ., one or more (including all) of M252Y, S254T, and T256E mutations according to the EU numbering convention).
  • the IgGl modified Fc may further comprise one or more of E430G, E430S, E430F, E430T, E345K, E345Q, E345R, E345Y, S440Y, and/or S440W according to EFT numbering.
  • Fc regions modified constant regions
  • An antibody dependent on binding to FcyR receptor to activate targeted receptors may lose its agonist activity if engineered to eliminate FcyR binding (see, e.g., Wilson el al. Cancer Cell 19: 101-113 (2011); Armour et al. Immunology 40:585-593 (2003); and White et al. Cancer Cell 27: 138-148 (2015)).
  • an anti- SIRPpl antibody of the present disclosure with the correct epitope specificity can activate the target antigen, with minimal adverse effects, when the antibody has an Fc domain from a human IgG2 isotype (CH1 and hinge region) or another type of Fc domain that is capable of preferentially binding the inhibitory FcyRIIB receptors, or a variation thereof.
  • the modified antibody Fc is an IgG2 modified Fc.
  • the IgG2 modified Fc comprises one or more modifications.
  • the IgG2 modified Fc comprises one or more amino acid substitutions (e.g., relative to a wild-type Fc region of the same isotype).
  • the one or more amino acid substitutions are selected from V234A (Alegre et al. Transplantation 57: 1537-1543 (1994); Xu et al. Cell Immunol, 200: 16-26 (2000)); G237A (Cole et al.
  • the Fc comprises an amino acid substitution at positions V234A and G237A according to EU numbering.
  • the Fc comprises an amino acid substitution at positions C219S or C220S according to EU numbering. In some embodiments of any of the IgG2 modified Fc, the Fc comprises an amino acid substitution at positions A330S and P331S according to EU numbering. In some embodiments of any of the IgG2 modified Fc, the Fc comprises an amino acid substitution at positions S267E and F328F according to EU numbering.
  • the Fc comprises a C127S amino acid substitution according to the EU numbering convention (White et al., (2015) Cancer Cell 27, 138- 148; Fightle et al. Protein Sci. 19:753-762 (2010); and WO 2008/079246).
  • the antibody has an IgG2 isotype with a Kappa light chain constant domain that comprises a C214S amino acid substitution according to the EU numbering convention (White et al. Cancer Cell 27: 138-148 (2015); Lightle et al. Protein Sci. 19:753-762 (2010); and WO 2008/079246).
  • the Fc comprises a C220S amino acid substitution according to the EU numbering convention.
  • the antibody has an IgG2 isotype with a Kappa light chain constant domain that comprises a C214S amino acid substitution according to the EU numbering convention.
  • the Fc comprises a C219S amino acid substitution according to the EU numbering convention.
  • the antibody has an IgG2 isotype with a Kappa light chain constant domain that comprises a C214S amino acid substitution according to the EU numbering convention.
  • the Fc includes an IgG2 isotype heavy chain constant domain l(CHl) and hinge region (White et al. Cancer Cell 27: 138-148 (2015)).
  • the IgG2 isotype CH1 and hinge region comprise the amino acid sequence of 118-230 according to EU numbering.
  • the antibody Fc region comprises a S267E amino acid substitution, a L328F amino acid substitution, or both, and/or a N297A or N297Q amino acid substitution according to the EU numbering convention.
  • the Fc further comprises one or more amino acid substitution at positions E430G, E430S, E430F, E430T, E345K, E345Q, E345R, E345Y, S440Y, and S440W according to EU numbering.
  • the Fc may further comprise one or more mutations to enhance the antibody half-life in human serum (e.g ., one or more (including all) of M252Y, S254T, and T256E mutations according to the EU numbering convention).
  • the Fc may further comprise A330S and P331S.
  • the Fc is an IgG2/4 hybrid Fc.
  • the IgG2/4 hybrid Fc comprises IgG2 aa 118 to 260 and IgG4 aa 261 to 447.
  • the Fc comprises one or more amino acid substitutions at positions H268Q, V309L, A33 OS, and P331S according to EU numbering.
  • the Fc comprises one or more additional amino acid substitutions selected from A330L, L234F; L235E, or P331S according to EU numbering; and any combination thereof.
  • the Fc comprises one or more amino acid substitutions at a residue position selected from C127S, L234A, L234F, L235A, L235E, S267E, K322A, L328F, A330S, P331S, E345R, E430G, S440Y, and any combination thereof according to EU numbering.
  • the Fc comprises an amino acid substitution at positions E430G, L243A, L235A, and P33 lS according to EU numbering.
  • the Fc comprises an amino acid substitution at positions E430G and P331 S according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at positions E430G and K322A according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at positions E430G, A33 OS, and P331S according to EFT numbering.
  • the Fc comprises an amino acid substitution at positions E430G, K322A, A33 OS, and P331S according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at positions E430G, K322A, and A33 OS according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at positions E430G, K322A, and P331S according to EFT numbering.
  • the Fc comprises an amino acid substitution at positions S267E and F328F according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at position C127S according to EFT numbering. In some embodiments of any of the IgGl and/or IgG2 modified Fc, the Fc comprises an amino acid substitution at positions E345R, E430G and S440Y according to EFT numbering.
  • the modified antibody Fc is an IgG4 modified Fc.
  • the IgG4 modified Fc comprises one or more modifications.
  • the IgG4 modified Fc comprises one or more amino acid substitutions (e.g relative to a wild-type Fc region of the same isotype).
  • the one or more amino acid substitutions are selected from F235A, G237A, S229P, F236E (Reddy et al.
  • the Fc may further comprise F235A, G237A, and E318 A according to the EFT numbering convention.
  • the Fc may further comprise S228P and F235E according to the EFT numbering convention.
  • the IgG4 modified Fc may further comprise S267E and F328F according to the EFT numbering convention.
  • the IgG4 modified Fc comprises may be combined with an S228P mutation according to the EU numbering convention (Angal et al. Mol Immunol. 30: 105-108 (1993)) and/or with one or more mutations described in (Peters et al. J Biol Chem. 287(29):24525-33 (2012)) to enhance antibody stabilization.
  • the IgG4 modified Fc may further comprise one or more mutations to enhance the antibody half-life in human serum (e.g., one or more (including all) of M252Y, S254T, and T256E mutations according to the EU numbering convention).
  • one or more mutations to enhance the antibody half-life in human serum e.g., one or more (including all) of M252Y, S254T, and T256E mutations according to the EU numbering convention.
  • the Fc comprises L235E according to EFT numbering. In certain embodiments of any of the IgG4 modified Fc, the Fc comprises one or more amino acid substitutions at a residue position selected from C127S, F234A, F235A, F235E, S267E, K322A, F328F, E345R, E430G, S440Y, and any combination thereof, according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc comprises an amino acid substitution at positions E430G, F243A, F235A, and P331S according to EFT numbering.
  • the Fc comprises an amino acid substitution at positions E430G and P331 S according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc comprises an amino acid substitution at positions E430G and K322A according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc comprises an amino acid substitution at position E430 according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc region comprises an amino acid substitution at positions E430G and K322A according to EFT numbering.
  • the Fc comprises an amino acid substitution at positions S267E and F328F according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc comprises an amino acid substitution at position C127S according to EFT numbering. In some embodiments of any of the IgG4 modified Fc, the Fc comprises an amino acid substitution at positions E345R, E430G and S440Y according to EFT numbering.
  • the antibody is a derivative.
  • derivative refers to a molecule that includes a chemical modification other than an insertion, deletion, or substitution of amino acids (or nucleic acids).
  • derivatives comprise covalent modifications, including, but not limited to, chemical bonding with polymers, lipids, or other organic or inorganic moieties.
  • a chemically modified antigen binding protein can have a greater circulating half-life than an antigen binding protein that is not chemically modified.
  • a chemically modified antigen binding protein can have improved targeting capacity for desired cells, tissues, and/or organs.
  • a derivative antigen binding protein is covalently modified to include one or more water soluble polymer attachments, including, but not limited to, polyethylene glycol, polyoxyethylene glycol, or polypropylene glycol. See, e.g., U.S. Pat. Nos. 4640835, 4496689, 4301144, 4670417, 4791192 and 4179337.
  • a derivative antigen binding protein comprises one or more polymer, including, but not limited to, monomethoxy -polyethylene glycol, dextran, cellulose, , copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, polyvinyl pyrrobdone, poly-l, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), poly-(N-vinyl pyrrolidone)-polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer,
  • polymer including, but not limited to, monomethoxy -polyethylene glycol, dextran, cellulose, , copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, polyvinyl pyrrobdone, poly-l, 3-dioxolane
  • polyoxyethylated polyols e.g., glycerol
  • polyvinyl alcohol as well as mixtures of such polymers.
  • a derivative is covalently modified with polyethylene glycol (PEG) subunits.
  • PEG polyethylene glycol
  • one or more water-soluble polymer is bonded at one or more specific position, for example at the amino terminus, of a derivative.
  • one or more water-soluble polymer is randomly attached to one or more side chains of a derivative.
  • PEG is used to improve the therapeutic capacity for an antigen binding protein.
  • PEG is used to improve the therapeutic capacity for a humanized antibody.
  • Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed“peptide mimetics” or“peptidomimetics.” Fauchere, J. Adv. Drug Res., 15:29 (1986); and Evans et al. J. Med. Chem., 30: 1229 (1987), which are incorporated herein by reference for any purpose. Such compounds are often developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides can be used to produce a similar therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biochemical property or
  • pharmacological activity such as human antibody
  • Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type e.g., D- lysine in place of L-lysine
  • D-amino acid of the same type e.g., D- lysine in place of L-lysine
  • constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation can be generated by methods known in the art (Rizo and Gierasch Ann. Rev. Biochem., 61 :387 (1992), incorporated herein by reference for any purpose); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide.
  • Drug conjugation involves coupling of a biological active cytotoxic (anticancer) payload or drug to an antibody that specifically targets a certain tumor marker (e.g. a polypeptide that, ideally, is only to be found in or on tumor cells).
  • a certain tumor marker e.g. a polypeptide that, ideally, is only to be found in or on tumor cells.
  • Antibodies track these proteins down in the body and attach themselves to the surface of cancer cells.
  • the biochemical reaction between the antibody and the target protein (antigen) triggers a signal m the tumor cell, which then absorbs or internalizes the antibody together with the cytotoxin.
  • the cytotoxic drug is released and kills the cancer. Due to this targeting, ideally the drug has lower side effects and gives a wider therapeutic window than other chemotherapeutic agents.
  • an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1, but does not bind to human SIRPa. In some embodiments, an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1, but does not bind to human SIRPy. In some embodiments, an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1, but does not bind to human SIRP 1 isoform 3. In some embodiments, an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1, but does not bind to mouse SIRP 1.
  • an anti-SIRP 1 antibody that binds to human SIRP 1 isoform 1, but does not bind to cynomolgus monkey SIRP 1. In some embodiments, an anti-SIRP 1 antibody is provided that binds to human SIRP 1 isoform 1, but does not bind to cynomolgus monkey SIRP 1 isoform 1.
  • binding of an anti-SIRP 1 antibody to an antigen may be determined using a ForteBio Octet® Red384 system (ForteBio, Menlo Park, CA), for example, at described in Example 1.
  • Antibody binding may be determined using, for example, a Fab fragment of the antibody (for monovalent binding) or a full-length antibody such as an IgG (for bivalent binding, or avidity).
  • An exemplary binding assay for full-length antibodies using a ForteBio Octet® Red384 system is as follows. Antibodies are loaded onto AHQ sensors. The loaded antibodies are then exposed to antigen, and the off-rate measured in assay buffer at various time intervals (such as 3 minutes).
  • Kinetics data may then be fit using a binding model in the data analysis software provided with the system.
  • an antigen-Fc may be loaded onto AHQ sensors and then exposed to the antibody Fab fragment. The off-rate is measured as above, and the data analyzed using the software provided with the system.
  • anti-SIRP 1 antibody that agonizes SIRP 1 activity on CD 14-positive monocytes in vitro and/or in vivo. In some embodiments, and anti-SIRP 1 antibody is provided that agonizes SIRP 1 activity on CD 14-positive monocytes in vitro. In some embodiments, and anti-SIRP 1 antibody is provided that agonizes SIRP 1 activity on CD 14- positive monocytes in vivo.
  • the anti-SIRP 1 antibody agonizes SIRP 1 activity on CDl4-positive monocytes in vitro in an assay in which the anti-SIRP 1 antibody is immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell.
  • a solid support such as a culture plate
  • a secondary anti-IgG antibody or bound by Fc gamma receptor on an accessory cell.
  • and anti-SIRP 1 antibody is provided that agonizes SIRP 1 activity on CD 14-positive monocytes in vivo.
  • a nonlimiting exemplary assay for determining whether an anti-SIRP 1 antibody agonizes SIRP 1 activity on CDl4-positive monocytes in vitro is described in Examples 3 and 4.
  • isolated monocytes are starved for a period of time, such as 4 hours, and then incubated with anti- SIRP 1 antibody, e.g., in the presence of anti-IgG antibody.
  • anti- SIRP 1 antibody e.g., in the presence of anti-IgG antibody.
  • cells are lysed and phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP12 is measured, for example, using an anti-phosphotyrosine and/or anti-phosphoserine antibody.
  • An increase in phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP12 is indicative of an increase in SIRP 1 activity (i.e., agonist of SIRP 1 activity).
  • monocytes may be incubated with anti-SRP 1 antibody in the presence of accessory cells that express Fc gamma receptors, such as B cells.
  • B cells such as B cells.
  • cells are lysed and phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP12 is determined as above.
  • An increase in phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP 12 is indicative of an increase in SIRP 1 activity (i.e., that the anti-SIRP 1 antibody is an agonist of SIRP 1 activity).
  • a nonlimiting exemplary assay to determine whether an anti-SIRP 1 antibody agonizes SIRP 1 activity on CD 14- positive monocytes in vivo comprises administering the anti-SIRP 1 antibody to a human SIRP 1 BAC transgenic C57BL/6 mouse, and isolating CDl4-positive monocytes, e.g., by FACS.
  • the monocytes are then lysed and phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP12 is measured as described above.
  • An increase in phosphorylation of one or more of Syk, ERK, AKT, SIRP 1 , and/or DAP12 is indicative of an increase in SIRP 1 activity (i.e., agonist of SIRP 1 activity).
  • an anti-SIRP 1 antibody that induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro and/or in vivo.
  • an anti-SIRP 1 antibody is provided that induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro.
  • an anti- SIRP 1 antibody is provided that induces or increases respiratory burst in immune cells, such as neutrophils and/or monocytes in vivo.
  • Nonlimiting exemplary in vitro assays for determining whether an anti-SIRP 1 antibody induces or increases respiratory burst in immune cells are described in Examples 5 and 6.
  • primary neutrophils are contacted with anti-SIRP 1 antibody and the production of reactive oxygen species (ROS) is detected using a general oxidative stress indicator, such as fluorescent dye CM-FEDCFDA.
  • ROS reactive oxygen species
  • the assay may be carried out using anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell.
  • a solid support such as a culture plate
  • a secondary anti-IgG antibody or bound by Fc gamma receptor on an accessory cell.
  • an anti-SIRP 1 antibody that induces or increases IL-8 expression in monocytes in vitro and/or in vivo. In some embodiments, an anti-SIRP 1 antibody is provided that induces or increases IL-8 expression in monocytes in vitro. In some embodiments, an anti-SIRP 1 antibody is provided that induces or increases IL-8 expression in monocytes in vivo.
  • a nonlimiting exemplary in vitro assay for determining whether an anti-SIRP 1 antibody induces or increases IL-8 expression in monocytes is described in Example 5.
  • primary monocytes may be stimulated with anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell, for example, overnight.
  • the supernatant may be collected to assay for IF-8 release.
  • a nonlimiting exemplary assay to determine whether an anti-SIRP 1 antibody induces or increases IF- 8 expression in monocytes in vivo comprises administering the anti-SIRP 1 antibody to human SIRP 1 BAC transgenic C57BF/6 mice and obtaining one or more blood samples.
  • Serum concentration of IF-8 may be determined using a commercial assay, such as the Duoset EFISA kit (R&D Systems).
  • an anti-SIRP 1 antibody that induces or increases TNFa expression in macrophages and/or dendritic cells in vitro and/or in vivo. In some embodiments, an anti-SIRP 1 antibody is provided that induces or increases TNFa expression in macrophages and/or dendritic cells in vitro. In some embodiments, an anti-SIRP 1 antibody is provided that induces or increases TNFa expression in macrophages and/or dendritic cells in vivo. A nonlimiting exemplary in vitro assay for determining whether an anti-SIRP 1 antibody induces or increases TNFa expression is described in Example 6.
  • monocyte-derived macrophages or dendritic cells may be stimulated with EPS in the presence of anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell, for example, overnight.
  • the supernatant may be collected to assay for TNFa release.
  • a nonlimiting exemplary assay to determine whether an anti-SIRP 1 antibody induces or increases TNFa expression in macrophages and/or dendritic cells in vivo comprises administering the anti-SIRP 1 antibody to human SIRP 1 BAC transgenic C57BF/6 mice along with EPS (such as 5 pg of FPS/mouse).
  • TNF-a concentration may be determined using a commercial assay, such as the Duoset EFISA kit (R&D Systems).
  • an anti-SIRP 1 antibody that induces or increases neutrophil-mediated phagocytosis, for example, of tumor cells in vitro and/or in vivo.
  • an anti-SIRP 1 antibody is provided that induces or increases neutrophil-mediated phagocytosis, for example, of tumor cells in vitro.
  • an anti-SIRP 1 antibody is provided that induces or increases neutrophil-mediated phagocytosis, for example, of tumor cells in vivo.
  • an anti-SIRP 1 antibody is provided that induces or increases neutrophil-mediated tumor cell clearance in vivo.
  • phagocytosis is described in Example 7.
  • primary neutrophils are contacted with anti- SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell.
  • Cancer cells such as Raji B cell lymphoma cells engineered to express luciferase, are co-cultured with the neutrophils and immobilized anti-SIRP 1 antibody in the presence of opsonizing antibody, such as anti-CD20 antibody. Viable Raji cells are quantified by measuring luciferase activity.
  • a nonlimiting exemplary assay to determine whether an anti-SIRP 1 antibody induces or increases neutrophil- mediated tumor cell clearance in vivo comprises injecting intravenously GFP-expressing B16F10 melanoma cells into human SIRP 1 BAC transgenic mice. Mice are then treated with the anti- SIRP 1 antibody with an opsonizing anti-gp75 antibody. Peripheral blood is collected, red blood cells are lysed, and white blood cells resuspended in FACS buffer.
  • Neutrophils are stained with anti- CD 1 lb and anti-Ly6G antibodies and analyzed by flow cytometry for the acquisition of green fluorescence signal from ingested tumor cells.
  • An increase in green fluorescence signal in the neutrophils compared to the same experiment with an isotype-matched control antibody indicates that anti-SIRP 1 antibody induces or increases neutrophil-mediated phagocytosis.
  • a further nonlimiting exemplary assay to determine whether an anti-SIRP 1 antibody induces or increases neutrophil-mediated tumor cell clearance in vivo uses a lung metastasis model, e.g., as follows. Fc receptor g-chain deficient C57BL/6 mice, which lack expression of activating FcyRs, are injected intravenously with B16F10 melanoma cells. Bone marrow neutrophils isolated from human SIRP 1 BAC transgenic mice are then injected intravenously with the anti-SIRP 1 antibody in combination with an opsonizing anti-gp75 antibody. After a period of time, mice are euthanized and lungs are harvested and fixed.
  • the number of metastatic tumor nodules with dark pigmentation are visually counted.
  • a reduction in the number of metastatic tumor nodules compared to the same experiment with an isotype-matched control antibody indicates that the anti-SIRPBl antibody induces or increases neutrophil-mediated tumor cell clearance in vivo.
  • an anti-SIRP 1 antibody is provided that increases TREM2 expression on macrophages in vitro and/or in vivo. In some embodiments, an anti-SIRP 1 antibody is provided that increases TREM2 expression on macrophages in vitro. In some embodiments, an anti-SIRP 1 antibody is provided that increases TREM2 expression on macrophages in vivo.
  • a nonlimiting exemplary in vitro assay for determining whether an anti-SIRP 1 antibody increases TREM2 expression on macrophages is described in Example 16.
  • monocyte-derived macrophages differentiated in culture with M-CSF are incubated with anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell.
  • TREM2 expression on the mcrophages is analyzed, e.g., by FACS analysis.
  • a nonlimiting exemplary assay to determine whether an anti- SIRP 1 antibody increases TREM2 expression on macrophages in vivo is as follows. Human SIRP 1 BAC transgenic C57BL/6 mice are challenged with aged sterile thioglycolate broth injected into the peritoneum. Following challenge, mice are treated with the anti-SIRP 1 antibody.
  • Macrophages are defined as CD1 lb+ Ly6C- F4/80+ cells, and may be analyzed by FACS to determine TREM2 expression levels.
  • an anti-SIRP 1 antibody that increases viability of macrophages in vitro and/or in vivo, alone and/or in combination with an agonist anti-TREM2 antibody. In some embodiments, an anti-SIRP 1 antibody is provided that increases viability of macrophages in vitro, alone and/or in combination with an agonist anti-TREM2 antibody. In some embodiments, an anti-SIRP 1 antibody is provided that increases viability of macrophages in vivo, alone and/or in combination with an agonist anti-TREM2 antibody. In some embodiments, an anti- SIRP 1 antibody is provided that increases viability of dendritic cells in vitro and/or in vivo.
  • an anti-SIRP 1 antibody is provided that increases viability of dendritic cells in vitro. In some embodiments, an anti-SIRP 1 antibody is provided that increases viability of dendritic cells in vivo.
  • Nonlimiting exemplary in vitro assays for determining whether an anti- SIRP 1 antibody increases viability of macrophages in vitro, alone or in combination with an agonist anti-TREM2 antibody, are described in Examples 17, 18, and 19.
  • monocyte- derived macrophages differentiated in culture with M-CSF are incubated with anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell, and with or without anti-TREM2 antibody.
  • Cell viability is measured, for example, using Cell Titer Glo kit (Promega), a reagent that produces a luminescence signal relative to ATP concentration in the sample.
  • a similar assay may be used to determine whether an anti-SIRP 1 antibody increases the viability of bone marrow-derived macrophages or dendritic cells in vitro.
  • bone marrow cells are cultured with M-CSF to differentiate macrophages, or with GM-CSF to differentiate dendritic cells.
  • the macrophages or dendritic cells are incubated with anti-SIRP 1 antibody immobilized on a solid support, such as a culture plate, or bound by a secondary anti-IgG antibody, or bound by Fc gamma receptor on an accessory cell, and with or without anti-TREM2 antibody.
  • a solid support such as a culture plate
  • a secondary anti-IgG antibody or bound by Fc gamma receptor on an accessory cell
  • Fc gamma receptor Fc gamma receptor on an accessory cell
  • cell viability is measured, for example, using Cell Titer Glo kit (Promega), a reagent that produces a luminescence signal relative to ATP concentration in the sample.
  • a nonlimiting exemplary assay to determine whether an anti- SIRP 1 antibody increases viability of macrophages in vivo is as follows.
  • mice are sacrificed, e.g., by CO2 asphyxiation, and peripheral blood is collected by cardiac puncture. Peritoneal macrophages are collected by lavage with PBS. Macrophage populations are counted by FACS by gating on CD1 lb+ Ly6C- F4/80+ cells in blood and peritoneum.
  • Anti-SIRP 1 antibodies of the present disclosure may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4816567.
  • isolated nucleic acids having a nucleotide sequence encoding any of the anti-SIRP 1 antibodies of the present disclosure are provided. Such nucleic acids may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VHof the anti-SIRP 1 antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is also provided.
  • the host cell comprises (e.g., has been transduced with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g., a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • Host cells of the present disclosure also include, without limitation, isolated cells, in vitro cultured cells, and ex vivo cultured cells.
  • Methods of making an anti-SIRP 1 antibody of the present disclosure include culturing a host cell of the present disclosure comprising a nucleic acid encoding the anti-SIRP 1 antibody, under conditions suitable for expression of the antibody.
  • the antibody is subsequently recovered from the host cell (or host cell culture medium).
  • nucleic acid encoding the anti-SIRP 1 antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable vectors comprising a nucleic acid sequence encoding any of the anti-SIRP 1 antibodies of the present disclosure, or cell-surface expressed fragments or polypeptides thereof polypeptides (including antibodies) described herein include, without limitation, cloning vectors and expression vectors. Suitable cloning vectors can be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art.
  • cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones comprising the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUCl8, pUCl9, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, ColEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen.
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells.
  • anti-SIRP 1 antibodies of the present disclosure may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microorganisms such as filamentous fungi or yeast
  • suitable cloning or expression hosts for antibody-encoding vectors including fungi and yeast strains whose glycosylation pathways have been“humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern (e.g., Gemgross Nat. Biotech. 22: 1409-1414 (2004); and Fi et al. Nat. Biotech. 24:210-215 (2006)).
  • Suitable host cells for the expression of glycosylated antibody can also be derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures can also be utilized as hosts (e.g., U.S. Patent Nos. 5959177, 6040498, 6420548, 7125978, and 6417429, describing PFANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al. J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HEFA); canine kidney cells (MDCK; buffalo rat liver cells (BRF 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al. Annals N. Y. Acad. Sci. 383:44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR- CHO cells (Urlaub et al. Proc. Natl. Acad. Sci.
  • compositions and/or pharmaceutical formulations comprising the anti-SIRP 1 antibodies of the present disclosure and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier preferably are nontoxic to recipients at the dosages and concentrations employed.
  • the antibodies described herein may be formulated into preparations in solid, semi-solid, liquid or gaseous forms. Examples of such formulations include, without limitation, tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • Pharmaceutically acceptable carriers can include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers of diluents, which are vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • the pharmaceutical composition can comprise formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • pharmaceutically acceptable carriers include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; e
  • amino acids such as g
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can comprise antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • Formulations may be optimized for retention and stabilization in the brain or central nervous system.
  • the agent When the agent is administered into the cranial compartment, it is desirable for the agent to be retained in the compartment, and not to diffuse or otherwise cross the blood brain barrier.
  • Stabilization techniques include cross-linking, multimerizing, or linking to groups such as polyethylene glycol, polyacrylamide, neutral protein carriers, etc. in order to achieve an increase in molecular weight.
  • Implants may be particles, sheets, patches, plaques, fibers, microcapsules and the like and may be of any size or shape compatible with the selected site of insertion.
  • Biodegradable polymeric compositions which may be employed may be organic esters or ethers, which when degraded result in physiologically acceptable degradation products, including the monomers. Anhydrides, amides, orthoesters or the like, by themselves or in combination with other monomers, may find use.
  • the polymers will be condensation polymers.
  • the polymers may be cross-linked or non-cross-linked.
  • polymers of hydroxyaliphatic carboxylic acids include homo- or copolymers, and polysaccharides. Included among the polyesters of interest are polymers of D-lactic acid, L-lactic acid, racemic lactic acid, glycolic acid, polycaprolactone, and combinations thereof.
  • polysaccharides of interest include calcium alginate, and functionalized celluloses, particularly carboxymethylcellulose esters characterized by being water insoluble, a molecular weight of about 5 kD to 500 kD, etc.
  • Biodegradable hydrogels may also be employed in the implants of the subject invention.
  • Hydrogels are typically a copolymer material, characterized by the ability to imbibe a liquid.
  • anti-SIRPpl antibodies of the present disclosure may be used for preventing, reducing risk, or treating diseases and disorders.
  • the anti- SIRP 1 antibodies provided herein have one or more activities selected from: agonizing SIRP 1 activity on CDl4-positive monocytes in vitro and/or in vivo, ⁇ inducing respiratory burst in immune cells, such as neutrophils and/or monocytes in vitro and/or in vivo, ⁇ inducing IL-8 expression in monocytes in vitro and/or in vivo; inducing TNFa expression in macrophages and/or dendritic cells in vitro and/or in vivo; inducing neutrophil-mediated phagocytosis, for example, of tumor cells in vitro and/or in vivo; increasing neutrophil-mediated tumor cell clearance in vivo; upregulating TREM2 expression on macrophages in vitro and/or in vivo; increasing viability of macrophages in vitro and/
  • the anti-SIRP 1 antibodies provided herein may be used for preventing, reducing risk, or treating dementia, frontotemporal dementia, Alzheimer’s disease, vascular dementia, mixed dementia, Creutzfeldt-Jakob disease, normal pressure hydrocephalus, amyotrophic lateral sclerosis, Huntington’s disease, Taupathy disease, Nasu-Hakola disease, stroke, acute trauma, chronic trauma, lupus, acute and chronic colitis, wound healing, Crohn's disease, inflammatory bowel disease, ulcerative colitis, obesity, Malaria, essential tremor, central nervous system lupus, Behcet's disease, Parkinson’s disease, dementia with Lewy bodies, multiple system atrophy, Shy-Drager syndrome, progressive supranuclear palsy, cortical basal ganglionic
  • the anti- SIRP 1 antibody is an agonist antibody.
  • provided herein are methods of preventing, reducing risk, or treating an individual having dementia, frontotemporal dementia, Alzheimer’s disease, vascular dementia, mixed dementia, Creutzfeldt-Jakob disease, normal pressure hydrocephalus, amyotrophic lateral sclerosis, Huntington’s disease, Taupathy disease, Nasu-Hakola disease, stroke, acute trauma, chronic trauma, lupus, acute and chronic colitis, wound healing, Crohn's disease, inflammatory bowel disease, ulcerative colitis, obesity, Malaria, essential tremor, central nervous system lupus, Behcet's disease, Parkinson’s disease, dementia with Lewy bodies, multiple system atrophy, Shy- Drager syndrome, progressive supranuclear palsy, cortical basal ganglionic degeneration, acute disseminated encephalomyelitis, granulomartous disorders, Sarcoidosis, diseases of aging, seizures, spinal cord injury, traumatic brain injury, age related macular degeneration
  • the anti-SIRP 1 antibody is an agonist of (e.g., induces or increases) SIRP 1 activity.
  • anti-SIRPpl antibodies of the present disclosure may also be used for inducing and/or promoting innate immune cell survival.
  • the present disclosure provides methods of inducing or promoting innate immune cell survival in an individual in need thereof, by administering to the individual a therapeutically effective amount of an agonist anti-SIRP 1 antibody of the present disclosure.
  • anti-SIRPpl antibodies of the present disclosure may also be used for inducing and/or promoting wound healing, such as after injury.
  • the wound healing may be colonic wound repair following injury.
  • the present disclosure provides methods of inducing or promoting wound healing in an individual in need thereof, by administering to the individual a therapeutically effective amount of an agonist anti-SIRP 1 antibody of the present disclosure.
  • a subject or individual is a mammal.
  • Mammals include, without limitation, domesticated animals (e.g ., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • the subject or individual is a human.
  • An antibody provided herein can be administered by any suitable means, including parenteral, intrapulmonary, intranasal, intralesional administration, intracerobrospinal, intracranial, intraspinal, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • Parenteral infusions include intramuscular, intravenous administration as a bolus or by continuous infusion over a period of time, intraarterial, intra-articular, intraperitoneal, or subcutaneous administration. In some embodiments, the administration is intravenous
  • the administration is subcutaneous.
  • Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies provided herein would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • 0.1 mg/kg- 10 mg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 pg/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg, or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermitently, e.g., every week or every three weeks (e.g., such that the patient receives from about two to about twenty, or e.g., about six doses of the antibody).
  • dosing frequency is three times per day, twice per day, once per day, once every other day, once weekly, once every two weeks, once every four weeks, once every five weeks, once every six weeks, once every seven weeks, once every eight weeks, once every nine weeks, once every ten weeks, or once monthly, once every two months, once every three months, or longer.
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • Dementia is a non-specific syndrome (i.e., a set of signs and symptoms) that presents as a serious loss of global cognitive ability in a previously unimpaired person, beyond what might be expected from normal ageing. Dementia may be static as the result of a unique global brain injury. Alternatively, dementia may be progressive, resulting in long-term decline due to damage or disease in the body. While dementia is much more common in the geriatric population, it can also occur before the age of 65. Cognitive areas affected by dementia include, without limitation, memory, attention span, language, and problem solving. Generally, symptoms must be present for at least six months to before an individual is diagnosed with dementia. [0193] Exemplary forms of dementia include, without limitation, frontotemporal dementia, Alzheimer's disease, vascular dementia, semantic dementia, and dementia with Lewy bodies.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat dementia.
  • administering an anti- SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having dementia.
  • Frontotemporal dementia is a condition resulting from the progressive deterioration of the frontal lobe of the brain. Over time, the degeneration may advance to the temporal lobe. Second only to Alzheimer's disease (AD) in prevalence, FTD accounts for 20% of pre-senile dementia cases.
  • the clinical features of FTD include memory deficits, behavioral abnormalities, personality changes, and language impairments (Cruts, M. & Van Broeckhoven, C., Trends Genet. 24: 186-194 (2008); Neary, D., et al., Neurology 51 : 1546-1554 (1998); Ratnavalli, E., Brayne, C., Dawson, K. & Hodges, J. R., Neurology 58: 1615-1621 (2002)).
  • FTD FTD
  • a substantial portion of FTD cases are inherited in an autosomal dominant fashion, but even in one family, symptoms can span a spectrum from FTD with behavioral disturbances, to Primary Progressive Aphasia, to Cortico-Basal Ganglionic Degeneration.
  • FTD like most neurodegenerative diseases, can be characterized by the pathological presence of specific protein aggregates in the diseased brain.
  • the first descriptions of FTD recognized the presence of intraneuronal accumulations of hyperphosphorylated Tau protein in neurofibrillary tangles or Pick bodies.
  • a causal role for the microtubule associated protein Tau was supported by the identification of mutations in the gene encoding the Tau protein in several families (Hutton, M., et al., Nature 393:702-705 (1998).
  • the majority of FTD brains show no accumulation of
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat FTD.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having FTD.
  • AD Alzheimer’s disease
  • AD is the most common form of dementia. There is no cure for the disease, which worsens as it progresses, and eventually leads to death. Most often, AD is diagnosed in people over 65 years of age. However, the less-prevalent early-ons et a/zheimer's can occur much earlier.
  • Alzheimer’s disease Common symptoms of Alzheimer’s disease include, behavioral symptoms, such as difficulty in remembering recent events; cognitive symptoms, confusion, irritability and aggression, mood swings, trouble with language, and long-term memory loss. As the disease progresses bodily functions are lost, ultimately leading to death. Alzheimer’s disease develops for an unknown and variable amount of time before becoming fully apparent, and it can progress undiagnosed for years.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat Alzheimer’s disease.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having
  • Nasu-Hakola disease which may alternatively be referred to as polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL)
  • PLOSL polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy
  • NHD disease course is generally divided into four stages: latent, osseous, early neurologic, and late neurologic. After a normal development during childhood (latent stage), NHD starts manifesting during adolescence or young adulthood (typical age of onset 20-30 years) with pain in the hands, wrists, ankles, and feet. Patients then start suffering from recurrent bone fractures due to polycystic osseous and
  • osteroporotic lesions in the limb bones osteroporotic lesions in the limb bones (osseous stage).
  • patients present with pronounced personality changes (e.g ., euphoria, lack of concentration, loss of judgment, and social inhibitions) characteristic of a frontal lobe syndrome. Patients also typically suffer from progressive memory disturbances. Epileptic seizures are also frequently observed.
  • patients progress to a profound dementia, are unable to speak and move, and usually die by the age of 50
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat Nasu-Hakola disease (NHD).
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having NHD.
  • Parkinson’s disease which may be referred to as idiopathic or primary parkinsonism, hypokinetic rigid syndrome (HRS), or paralysis agitans, is a neurodegenerative brain disorder that affects motor system control. The progressive death of dopamine-producing cells in the brain leads to the major symptoms of Parkinson’s. Most often, Parkinson’s disease is diagnosed in people over 50 years of age. Parkinson’s disease is idiopathic (having no known cause) in most people.
  • Symptoms of Parkinson’s disease include, without limitation, tremors of the hands, arms, legs, jaw, and face, muscle rigidity in the limbs and trunk, slowness of movement (bradykinesia), postural instability, difficulty walking, neuropsychiatric problems, changes in speech or behavior, depression, anxiety, pain, psychosis, dementia, hallucinations, and sleep problems.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat Parkinson’s disease.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having
  • amyotrophic lateral sclerosis or motor neuron disease or Lou Gehrig's disease are used interchangeably and refer to a debilitating disease with varied etiology characterized by rapidly progressive weakness, muscle atrophy and fasciculations, muscle spasticity, difficulty speaking (dysarthria), difficulty swallowing (dysphagia), and difficulty breathing
  • progranulin play a role in ALS (Schymick, JC et al, (2007) J Neurol Neurosurg Psychiatry. ;78:754-6) and protects again the damage caused by ALS causing proteins such as TDP-43 (Laird, AS et al, (2010). PLoS ONE 5: el3368). It was also demonstrated that pro-NGF induces p75 mediated death of oligodendrocytes and corticospinal neurons following spinal cord injury (Beatty et al, Neuron (2002), 36, pp. 375-386; Giehl et al, Proc. Natl. Acad. Sci USA (2004), 101, pp 6226-30).
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat ALS.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having ALS.
  • Huntington’s disease is an inherited neurodegenerative disease caused by an autosomal dominant mutation in the Huntingtin gene (HTT). Expansion of a cytokine-adenine- guanine (CAG) triplet repeat within the Huntingtin gene results in production of a mutant form of the Huntingtin protein (Htt) encoded by the gene. This mutant Huntingtin protein (mHtt) is toxic and contributes to neuronal death. Symptoms of Huntington’s disease most commonly appear between the ages of 35 and 44, although they can appear at any age.
  • Symptoms of Huntington’ s disease include, without limitation, motor control problems, jerky, random movements (chorea), abnormal eye movements, impaired balance, seizures, difficulty chewing, difficulty swallowing, cognitive problems, altered speech, memory deficits, thinking difficulties, insomnia, fatigue, dementia, changes in personality, depression, anxiety, and compulsive behavior.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat Huntington’s disease (HD).
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having HD.
  • Taupathy diseases are a class of neurodegenerative disease caused by aggregation of the microtubule-associated protein tau within the brain.
  • AD Alzheimer’s disease
  • NFTs insoluble neurofibrillary tangles
  • Taupathy diseases and disorders include progressive supranuclear palsy, dementia pugilistica (chromic traumatic encephalopathy), Frontotemporal dementia and parkinsonism linked to chromosome 17, Lytico-Bodig disease (Parkinson-dementia complex of Guam), Tangle-predominant dementia, Ganglioglioma and gangliocytoma, Meningioangiomatosis, Subacute sclerosing panencephalitis, lead encephalopathy, tuberous sclerosis, Hallervorden-Spatz disease, lipofuscinosis, Pick’s disease, corticobasal degeneration, Argyrophilic grain disease (AGD), Huntington’s disease, frontotemporal dementia, and frontotemporal lobar degeneration.
  • ATD Argyrophilic grain disease
  • Huntington Huntington’s disease
  • frontotemporal dementia frontotemporal lobar degeneration.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat Taupathy disease.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having Taupathy disease.
  • MS Multiple sclerosis
  • disseminated sclerosis can also be referred to as disseminated sclerosis or
  • MS is an inflammatory disease in which the fatty myelin sheaths around the axons of the brain and spinal cord are damaged, leading to demyelination and scarring as well as a broad spectrum of signs and symptoms.
  • MS affects the ability of nerve cells in the brain and spinal cord to communicate with each other effectively. Nerve cells communicate by sending electrical signals called action potentials down long fibers called axons, which are contained within an insulating substance called myelin.
  • axons which are contained within an insulating substance called myelin.
  • myelin an insulating substance
  • Symptoms of MS include, without limitation, changes in sensation, such as loss of sensitivity or tingling; pricking or numbness, such as hypoesthesia and paresthesia; muscle weakness; clonus; muscle spasms; difficulty in moving; difficulties with coordination and balance, such as ataxia; problems in speech, such as dysarthria, or in swallowing, such as dysphagia; visual problems, such as nystagmus, optic neuritis including phosphenes, and diplopia; fatigue; acute or chronic pain; and bladder and bowel difficulties; cognitive impairment of varying degrees; emotional symptoms of depression or unstable mood; Uhthoff s phenomenon, which is an exacerbation of extant symptoms due to an exposure to higher than usual ambient temperatures; and Lhermitte's sign, which is an electrical sensation that runs down the back when bending the neck.
  • administering an anti-SIRP 1 antibody of the present disclosure can prevent, reduce the risk, and/or treat multiple sclerosis.
  • administering an anti-SIRP 1 antibody may induce one or more SIRP 1 activities in an individual having multiple sclerosis.
  • Yet further aspects of the present disclosure provide methods for preventing, reducing risk, or treating an individual having cancer, comprising administering to the individual a therapeutically effective amount of an isolated anti-SIRP 1 antibody of the present disclosure.
  • Any of the isolated antibodies of the present disclosure may be used in these methods.
  • the isolated antibody is an agonist antibody of the present disclosure.
  • the tumor microenvironment is known to contain a heterogeneous immune infiltrate, which includes T lymphocytes, macrophages and cells of myeloid/granulocytic lineage.
  • a heterogeneous immune infiltrate which includes T lymphocytes, macrophages and cells of myeloid/granulocytic lineage.
  • M2-macrophages in tumors is associated with poor prognosis.
  • therapies that reduce the number of these cells in the tumor are showing beneficial effects in preclinical models and early stage clinical studies.
  • a seminal preclinical study has also shown synergies between drugs that target tumor-associated macrophages (e.g.
  • a cancer to be prevented or treated by the methods of the present disclosure includes, without limitation, squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer, small-cell lung cancer, non-small cell lung cancer, squamous non-small cell lung cancer (NSCLC), adenocarcinoma of the lung, squamous carcinoma of the lung, non-squamous NSCLC, glioma, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, gastric or stomach cancer including gastrointestinal cancer and gastrointestinal stromal cancer, renal cancer (e.g., epithelial squamous cell carcinoma), lung cancer, small-cell lung cancer, non-small cell lung cancer, squamous non-small cell lung cancer (NSCLC), adenocarcinoma of the lung, squamous carcinoma of the lung, non-squamous NSCLC, glioma, cancer of the peritoneum, hepatocellular
  • ovarian cancer clear cell carcinoma
  • kidney cancer e.g., renal cell carcinoma (RCC)
  • prostate cancer e.g. hormone refractory prostate adenocarcinoma
  • thyroid cancer neuroblastoma, pancreatic cancer, glioblastoma (glioblastoma multiforme), cervical cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer (or carcinoma), gastric cancer, germ cell tumor, pediatric sarcoma, sinonasal natural killer, melanoma (e.g., metastatic malignant melanoma, such as cutaneous or intraocular malignant melanoma), bone cancer, skin cancer, uterine cancer, cancer of the anal region, testicular cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, cancer of the esophagus, cancer
  • lymphoplasmacytoid lymphoma monocytoid B-cell lymphoma, mucosa-associated lymphoid tissue (MALT) lymphoma, anaplastic (e.g., Ki 1+) large-cell lymphoma, adult T-cell lymphoma/leukemia, mantle cell lymphoma, angio immunoblastic T-cell lymphoma, angiocentric lymphoma, intestinal T- cell lymphoma, primary mediastinal B-cell lymphoma, precursor T-lymphoblastic lymphoma, T- lymphoblastic; and lymphoma/leukemia (T-Lbly/T-ALL), peripheral T-cell lymphoma,
  • lymphoblastic lymphoma post-transplantation lymphoproliferative disorder, true histiocytic lymphoma, primary central nervous system lymphoma, primary effusion lymphoma, lymphoblastic lymphoma (LBL), hematopoietic tumors of lymphoid lineage, acute lymphoblastic leukemia, diffuse large B-cell lymphoma, Burkitt's lymphoma, follicular lymphoma, diffuse histiocytic lymphoma (DHL), immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, cutaneous T-cell lymphoma (CTLC) (also called mycosis fungoides or Sezary syndrome), and lymphoplasmacytic lymphoma (LPL) with Waldenstrom's macroglobulinemia; myelomas, such as IgG myeloma, light chain myeloma, nonsecretory myeloma, smolder
  • T-cell and B-cell tumors including but not limited to T-cell disorders such as T-prolymphocytic leukemia (T-PLL), including of the small cell and cerebriform cell type; large granular lymphocyte leukemia (LGL) preferably of the T-cell type; a/d T-NHL heptasyllabic lymphoma; peripheral/post-thymic T cell lymphoma (pleomorphic and immunoblastic subtypes); angiocentric (nasal) T-cell lymphoma; cancer of the head or neck, renal cancer, rectal cancer, cancer of the thyroid gland; acute myeloid lymphoma, as well as any combinations of said cancers.
  • An anti-SIRP 1 antibody of the present disclosure may also be used to treat metastatic cancer.
  • the cancer is selected from sarcoma, bladder cancer, brain cancer, breast cancer, colon cancer, rectal cancer, endometrial cancer, kidney cancer, renal pelvis cancer, leukemia, lung cancer, small cell lung cancer, melanoma, lymphoma, pancreatic cancer, prostate cancer, ovarian cancer, and fibrosarcoma.
  • the cancer is triple-negative breast carcinoma.
  • the cancer may be an early stage cancer or a late stage cancer.
  • the cancer may be a primary tumor.
  • the cancer may be a metastatic tumor at a second site derived from any of the above types of cancer.
  • the cancer is selected from glioblastoma multiforme; renal clear cell carcinoma; adrenocortical carcinoma; bladder urothelial carcinoma; diffuse large B-cell lymphoma; lung adenocarcinoma; pancreatic adenocarcinoma, renal cell cancer, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, indolent B cell lymphoma, aggressive B cell lymphoma, T cell lymphoma, acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), multiple myeloma,
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myeloid leukemia
  • myelodysplastic syndromes myeloproliferative neoplasms
  • breast invasive carcinoma cervical squamous cell carcinoma
  • endocervical adenocarcinoma endocervical adenocarcinoma
  • cholangiocarcinoma colon
  • adenocarcinoma diffuse large B-cell lymphoma, esophageal carcinoma, head and neck squamous cell carcinoma, kidney chromophobe, renal papillary cell carcinoma, lower grade glioma, hepatocellular carcinoma, lung squamous cell carcinoma, mesothelioma, ovarian serous
  • cystadenocarcinoma pancreatic adenocarcinoma, pheochromocytoma and paraganglioma, prostate adenocarcinoma, rectal adenocarcinoma, cutaneous melanoma, stomach adenocarcinoma, testicular germ cell tumors, thyroid carcinoma, thymoma, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the present disclosure provides methods of preventing, reducing risk, or treating an individual having cancer, by administering to the individual a therapeutically effective amount of an anti-SIRP 1 antibody of the present disclosure.
  • the present disclosure provides methods of preventing, reducing risk, or treating an individual having cancer, wherein the individual is refractory to checkpoint inhibitor therapy, by administering to the individual a therapeutically effective amount of an anti- SIRP 1 antibody of the present disclosure.
  • an anti-SIRP 1 antibody of the present disclosure may be administered in conjunction with a therapeutic agent that acts as a checkpoint inhibitor.
  • the method further includes administering to the individual at least one antibody that specifically binds to an inhibitory immune checkpoint molecule, and/or another standard or investigational anti-cancer therapy.
  • the inhibitory checkpoint molecule is selected from PD1, PDL1, CD40, 0X40, ICOS, CD28, CD137/4-1BB, CD27, GITR, CTLA4, PD- L2, B7-H3, B7-H4, HVEM, LIGHT, BTLA, CD30, TIGIT, VISTA, KIR, GAL9, TIM1, TIM3, TIM4, A2AR, LAG3, DR-5, CD2, CD5, CD39, and CD73.
  • the therapeutic agent is an antibody to a checkpoint inhibitor selected from Dl, PDL1, CD40, 0X40, ICOS, CD28, CD137/4-1BB, CD27, GITR, CTLA4, PD-L2, B7-H3, B7-H4, HVEM, LIGHT, BTLA, CD30, TIGIT, VISTA, KIR, GAL9, TIM1, TIM3, TIM4, A2AR, LAG3, DR-5, CD2, CD5, CD39, or CD73.
  • the at least one antibody that specifically binds to an inhibitory checkpoint molecule is administered in combination with the anti-SIRP 1 antibody of the present disclosure.
  • a combination of antibodies to directed against checkpoint inhibitors is administered in conjunction with an anti-SIRP 1 antibody of the present invention.
  • an anti-SIRP 1 antibody of the present disclosure may be administered in conjunction with at least one agonistic antibody that specifically binds to a stimulatory checkpoint protein, e.g., an agonist anti-CD40 antibody, an agonist anti-OX40 antibody, an agonist anti-ICOS antibody, an agonist anti-CD28 antibody, an agonistic anti-TREMl antibody, an agonistic anti-TREM2 antibody, an agonist anti-CD 137/4- 1BB antibody, an agonist anti-CD27 antibody, an agonist anti-glucocorticoid-induced TNLR-related protein GITR antibody, an agonist anti-CD30 antibody, an agonist anti -BTLA antibody, an agonist anti -HVEM antibody, an agonist anti-CD2 antibody, an agonist anti-CD5 antibody, and any combination thereof.
  • a stimulatory checkpoint protein e.g., an agonist anti-CD40 antibody, an agonist anti-OX40 antibody, an agonist anti-ICOS antibody, an agonist anti-CD
  • the method further includes administering to the individual at least one antibody that specifically binds to an inhibitory immune checkpoint molecule, and/or another standard or investigational anti-cancer therapy.
  • the at least one antibody that specifically binds to an inhibitory checkpoint molecule is administered in combination with the anti- SIRP 1 antibody of the present disclosure.
  • the at least one antibody that specifically binds to an inhibitory checkpoint molecule is selected from an anti-PD-Ll antibody, an anti-CTLA4 antibody, an anti-PD-L2 antibody, an anti -PD- 1 antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, and anti -HVEM antibody, an anti- B- and T-lymphocyte attenuator (BTLA) antibody, an anti- Killer inhibitory receptor (KIR) antibody, an anti-GAL9 antibody, an anti-TIM3 antibody, an anti-A2AR antibody, an anti-LAG-3 antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, and any combination thereof.
  • BTLA B- and T-lymphocyte attenuator
  • KIR killer inhibitory receptor
  • the standard or investigational anti-cancer therapy is one or more therapies selected from radiotherapy, cytotoxic chemotherapy, targeted therapy, imatinib (Gleevec®), trastuzumab (Herceptin®), adoptive cell transfer (ACT), chimeric antigen receptor T cell transfer (CAR-T), vaccine therapy, and cytokine therapy.
  • the method further includes administering to the individual at least one antibody that specifically binds to an inhibitory cytokine.
  • the at least one antibody that specifically binds to an inhibitory cytokine is administered in combination with the anti-SIRP 1 antibody of the present disclosure.
  • the at least one antibody that specifically binds to an inhibitory cytokine is selected from an anti-CCL2 antibody, an anti-CSF-l antibody, an anti-IL-2 antibody, and any combination thereof.
  • the method further includes administering to the individual at least one agonistic antibody that specifically binds to a stimulatory immune checkpoint protein.
  • the at least one agonistic antibody that specifically binds to a stimulatory checkpoint protein is administered in combination with the anti-SIRP 1 antibody of the present disclosure.
  • the at least one agonistic antibody that specifically binds to a stimulatory checkpoint protein is selected from an agonist anti-CD40 antibody, an agonist anti-OX40 antibody, an agonist anti-ICOS antibody, an agonist anti-CD28 antibody, an agonist anti-CD 137/4- 1BB antibody, an agonist anti-CD27 antibody, an agonist anti-glucocorticoid-induced TNFR-related protein GITR antibody, and any combination thereof.
  • an anti-SIRP 1 antibody of the present invention is administered in combination with radiation therapy and/or a chemotherapeutic agent.
  • Chemotherapeutic agents include, for example, the following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (methotrexate, pemetrexed, mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones, eribulin
  • antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkylsulfonates (busulfan), nitrosoureas (carmustine (BCNU) and analogs, streptozocin), triazenes (dacarbazine (DTIC)); antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkylsulfonates (busulfan), nitrosoureas (carmustine (BCNU) and analogs, streptozocin), triazen
  • antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes
  • pomalidomide and growth factor inhibitors
  • vascular endothelial growth factor (VEGF) inhibitors such as ziv-aflibercept; fibroblast growth factor (FGF) inhibitors
  • IAP apoptosis protein
  • birinapant histone deacetylase (HD AC) inhibitors
  • vorinostat romidepsin, chidamide, panobinostat, mocetinostat, abexinostat, belinostat, entinostat, resminostat, givinostat, quisinostat, SB939)
  • proteasome inhibitors ixazomib
  • angiotensin receptor blocker nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab, panitumumab, pertuzumab, cetuximab, adalimumab, golimumab, infliximab, rituximab
  • rovelizumab ruplizumab, ustekinumab, visilizumab, gemtuzumab ozogamicin, brentuximb vedotin
  • chimeric antigen receptors cell cycle inhibitors (flavopiridol, roscovitine, bryostatin-l) and differentiation inducers (tretinoin); mTOR inhibitors, topoisomerase inhibitors (doxorubicin
  • iriamycin amsacrine, camptothecin, daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, irinotecan (CPT-l l) and mitoxantrone, topotecan, irinotecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); PARP inhibitors (niraparib, olaparib); focal adhesion kinase (FAK) inhibitors (defactinib (VS-6063), VS- 4718, VS-6062, GSK2256098); growth factor signal transduction kinase inhibitors (cediranib, galunisertib, rociletinib, vandetanib, afatinib, EGF816, AZD4547); c
  • a chemotherapeutic agent is a B-Raf inhibitor, a MEK inhibitor, a VEGF inhibitor, a VEGFR inhibitor, a tyrosine kinase inhibitor, an anti-mitotic agent, or any combination thereof.
  • an anti-SIRP 1 antibody of the present disclosure is administered in combination with adoptive cell transfer (ACT) therapy, chimeric antigen receptor T cell transfer (CAR-T) therapy, vaccine therapy, and/or cytokine therapy.
  • ACT adoptive cell transfer
  • CAR-T chimeric antigen receptor T cell transfer
  • an anti-SIRP 1 antibody of the present disclosure is administered in combination with at least one antibody that specifically binds to an inhibitory cytokine, e.g., an inhibitory cytokine such as an anti-CCL2 antibody, an anti-CSF-l antibody, or an anti-IL-2 antibody.
  • an inhibitory cytokine such as an anti-CCL2 antibody, an anti-CSF-l antibody, or an anti-IL-2 antibody.
  • an anti-SIRP 1 antibody of the present disclosure is administered in combination with at least one stimulatory cytokine.
  • the at least one stimulatory cytokine is selected from IFN- a4, IFN-b, IL- I b, TNF-a, IL-6, IL-8, CRP, IL-20 family members, LIF, IFN-g, OSM, CNTF, GM- CSF, IL-l l, IL-12, IL-15, IL-17, IL-18, IL-23, CXCL10, IL-33, MCP-l, MIP-l-beta, and any combination thereof.
  • any of the anti-SIRl ⁇ l antibodies provided herein is useful for detecting the presence of bIKRbI in a sample or an individual.
  • the term “detecting” as used herein encompasses quantitative or qualitative detection.
  • methods of using the antibodies of this disclosure for diagnostic purposes such as the detection of bIKRbI in an individual or in tissue samples derived from an individual.
  • the individual is a human.
  • the detection method may involve quantification of the antigen-bound antibody.
  • Antibody detection in biological samples may occur with any method known in the art, including
  • the antibody is radiolabeled, for example with 18 F and subsequently detected utilizing micro-positron emission tomography analysis.
  • Antibody-binding may also be quantified in a patient by non-invasive techniques such as positron emission tomography (PET), X-ray computed tomography, single photon emission computed tomography (SPECT), computed tomography (CT), and computed axial tomography (CAT).
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • CT computed tomography
  • CAT computed axial tomography
  • Article of manufacture may include one or more containers comprising an antibody described herein.
  • Containers may be any suitable packaging including, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like.
  • the containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • the kits may further include a second agent.
  • the second agent is a pharmaceutically-acceptable buffer or diluting agent including, but not limited to, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate- buffered saline phosphate- buffered saline
  • Ringer's solution phosphate- buffered saline
  • dextrose solution a pharmaceutically active agent.
  • the second agent is a pharmaceutically active agent described herein.
  • the article of manufactures further include instructions for use in accordance with the methods of this disclosure.
  • the instructions generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • these instructions comprise a description of administration of the isolated antibody of the present disclosure (e.g an anti-SIRP 1 antibody described herein) to prevent, reduce risk, or treat an individual having a disease, disorder, or injury selected from dementia, frontotemporal dementia, Alzheimer’s disease, vascular dementia, mixed dementia, Creutzfeldt-Jakob disease, normal pressure hydrocephalus, amyotrophic lateral sclerosis, Huntington’s disease, taupathy disease, Nasu-Hakola disease, stroke, acute trauma, chronic trauma, lupus, acute and chronic colitis, rheumatoid arthritis, wound healing, Crohn's disease, inflammatory bowel disease, ulcerative colitis, obesity, malaria, essential tremor, central nervous system lupus, Behcet
  • Example 1 Production identification and characterization of agonist anti-SIRPB 1 antibodies
  • Human SIRP 1 contains a signal peptide located at amino residues 1-29 of SEQ ID NO: 1; an extracellular immunoglobulin-like variable-type (IgV) domain located at amino residues 30-136 of SEQ ID NO: 1; two immunoglobulin-like constant (IgC) domains located at amino acids 147-246 and 253-347 SEQ ID NO: 1; a transmembrane domain located at amino residues 372-392 of SEQ ID NO: 1; and an intracellular domain located at amino residues 393-398 of SEQ ID NO: 1.
  • IgV extracellular immunoglobulin-like variable-type domain
  • IgC immunoglobulin-like constant domains
  • the human SIRPB 1 amino acid sequence comprises a lysine residue (aa380) within the transmembrane domain that interacts with an aspartic acid in DAP 12, a key adaptor protein that transduces signaling from SIRPB 1 , as well as TREM1, TREM2, and other related IgV family members.
  • a BLAST analysis of human SIRPB 1 identified multiple isoforms possibly derived from alternative splicing. Among these transcript variants, SIRPB 1 isoform 3 shares the most sequence identity to SIRPB 1 isoform 1 (FIG. 1A).
  • Human SIRPB 1 is also related to human SIRPa. An alignment of the amino acid sequences of human SIRPB 1 and human SIRPa was generated by 2- way blast (FIG. IB) The extracellular region of both receptors share 85% sequence identity. In contrast, human SIRPB 1 displays poorer homology to mouse SIRPB 1 (FIG. 2) suggesting that the SIRP gene loci in both species experience divergent selection pressure.
  • extracellular IgV domain (amino acid residues 30-136 of SEQ ID NO: 1) can be generated using mouse hybridoma technology, phage display technology, and yeast-based platform technology. Antibodies are screened for their ability to bind cells that express SIRPB 1 and for their ability to activate SIRPB 1 signaling, activity, and function in cells and in a whole animal in vivo as described in Examples 2-20 below. For example, agonistic anti-SIRP 1 antibodies can be produced that target the IgV domain (amino acid residues 30-136). By analogy to SIRPa, the IgV domain of SIRP 1 is predicted bind to an unknown ligand(s), and through multimerization of receptor, lead to activation. Production of His-tagged and Fc-coniugated human SIRPa and human SIRPB 1 IgV domains
  • SIRPa and SIRPB 1 monomer antigens were prepared by fragmenting a SIRPa/b 1 Fc fusion antigen with modified hinge region (Fynaugh et al., MAbs. 2013 Oct;5(5):64l-45) with FabRICATOR (IdeS) protease (Genovis, Cat # A2-FR2-1000), followed by Protein A affinity purification to remove undigested Fc fusion protein and SEC to remove aggregated monomer.
  • yeast cells (10 9 cells/library, total density -10 10 ) were incubated with 3 ml of 10 nM biotinylated SIRPB 1 -Fc fusion antigen or 100 nM biotinylated SIRPB 1 monomer antigen for 15 min at room temperature in FACS wash buffer PBS with 0.1% BSA. After washing once with 50 ml ice-cold wash buffer, the cell pellet was resuspended in 40 mF wash buffer, and 500 pL Streptavidin MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany. Cat # 130-048-101) were added to the yeast and incubated for 15 min at 4°C.
  • yeast cells were pelleted, resuspended in 5 mF wash buffer, and loaded onto a MACS FS column (Miltenyi Biotec, Bergisch Gladbach, Germany. Cat.#l30-042-40l). After the 5 mF was loaded, the column was washed 3 times with 3 ml FACS wash buffer. The column was then removed from the magnetic field, and the yeast were eluted with 5 mL of growth media and then grown overnight. The following four rounds of sorting were performed using flow cytometry.
  • Yeast were then washed twice and stained with goat anti -human F(ab')2 kappa-FITC diluted 1 : 100 (Southern Biotech, Birmingham, Alabama, Cat# 2062-02) and either streptavidin-Alexa Fluor 633 (Life Technologies, Grand Island, NY, Cat # S21375) diluted 1 :500, or Extravidin-phycoerythrin (Sigma-Aldrich, St Louis, Cat # E40l l) diluted 1 :50, secondary reagents for 15 min at 4°C. After washing twice with ice-cold wash buffer, the cell pellets were resuspended in 0.4 mL wash buffer and transferred to strainer-capped sort tubes.
  • Sorting was performed using a FACS ARIA sorter (BD Biosciences) and sort gates were determined to select only SIRP 1 binding clones. In the following selection round approximately 2 x 10 7 yeast were prepared as above but incubation was with a polyspecific reactivity reagent to conduct a negative sort to decrease poly specific binders (Xu et al., PEDS. 2013 Oct;26(l0):663-70), and binders to control protein, HIS-tagged human SIRPa monomer. The next round utilized labeling with 10 nM human SIRP 1 -Fc fusion antigens and 100 nM human SIRP 1 monomer antigen. After the final round of sorting, yeast cells were plated and individual colonies were picked for characterization. The final round utilized labeling with 100 nM and 10 nM human SIRP 1 monomer antigen.
  • Heavy chains from the second and fourth FACS sorting selection round outputs were used to prepare light chain diversification libraries used for additional selections.
  • the first selection round utilized Miltenyi MACs beads and labeling with 10 nM human SIRP 1 -Fc fusion antigen.
  • Four rounds of FACS sorting followed. The first round used 100 nM human SIRP 1 monomer antigen.
  • the second FACS round was a negative sort to decrease binding to reagent binders, polyspecific binders, and binders to control protein human SIRPa HIS tagged monomer.
  • the last two rounds utilized human SIRP 1 monomer titration (100 nM, 10 nM, and 1 nM) to select highest affinity binders, 100 nM human SIRPa monomer, and competition with control AM4-5 antibody, which binds to SIRP 1 IgV domain, to assess competitor representation in the enriched population.
  • human SIRP 1 monomer titration 100 nM, 10 nM, and 1 nM
  • competition with control AM4-5 antibody which binds to SIRP 1 IgV domain
  • Yeast clones were grown to saturation and then induced for 48 h at 30°C with shaking. After induction, yeast cells were pelleted and the supernatants were harvested for purification. IgGs were purified using a Protein A column and eluted with acetic acid, pH 2.0. Fab fragments were generated by papain digestion and purified over CaptureSelect IgG-CHl affinity matrix (LifeTechnologies, Cat # 1943200250).
  • affinities of the anti-SIRP 1 antibodies were determined by measuring their dissociation constants (KD) using a ForteBio Octet ® Red384 system (ForteBio, Menlo Park, CA), performed generally as previously described (Estep et al, MAbs. 2013 Mar-Apr;5(2):270-8). Briefly, Octet affinity measurements were performed by loading IgGs on-line onto AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 min and then monitored on-line for 60 seconds for baseline establishment.
  • KD dissociation constants
  • sensors with loaded IgGs were exposed to 100 nM antigen (human SIRPa domain 1 (Dl) Fc fusion or SIRP 1 domain 1 (Dl) Fc fusion) for 3 min, and then transferred to assay buffer for 3 min for off-rate measurement. Additional avid binding was determined by loading biotinylated SIRP 1 monomer on SA sensors and exposing to 100 nM IgG in solution. Monovalent binding measurements were obtained by loading human SIRPa- or SIRP 1 -Fc fusion antigens onto AHQ sensor, followed by exposure to 100 nM anti-SIRP 1 antibody Fab.
  • the final set of anti-SIRP 1 antibodies were selected based on antigen binding affinities. Antibodies that were positive for binding to human SIRP 1 were tested for cross-reactivity with human SIRPa. The bin category of each of the antibodies are listed below in Table 2. In Table 2, “ND” refers to antibodies for which the Bin category was not determined;“NB” refers to antibodies for which there was no binding to indicated antigen;“PF” refers to antibodies for which antigen binding kinetics showed poor fit to 1 : 1 binding model. No detectable binding to mouse SIRP 1 Dl- Fc, human SIRPa Dl-Fc, or mouse SIRPa Dl-Fc was observed for any of the anti-SIRP i antibodies. Table 2: Biochemical Characterization of anti-SIRPpi antibodies
  • the amino acid sequences encoding the light chain variable domains and the heavy chain variable domains of the antibodies were determined.
  • the EU or Kabat sequences of the antibodies are set forth in Tables 3-6, as follows.
  • the EU or Kabat light chain HVR sequences of the antibodies are set forth in Table 3.
  • the EU or Kabat heavy chain HVR sequences of the antibodies are set forth in Table 4.
  • the EU or Kabat light chain framework (FR) sequences of the antibodies are set forth in Table 5.
  • the EU or Kabat heavy chain framework (FR) sequences of the antibodies are set forth in Table 6.
  • MFI fluorescence intensity
  • Table 7 shows the mean fluorescence intensity (MFI) values of anti-SIRPB 1 antibodies binding to a Chinese hamster ovary (CHO) cell line expressing low levels of recombinant human SIRPB 1.
  • the human IgGl isotype control established the background fluorescence signal set to 1.
  • 32 clones bound to cells with an MFI > 2-fold over background.
  • the anti-SIRPB 1 antibodies were also screened for surface binding to CHO cells overexpressing recombinant mouse SIRPB 1. As expected, none of the test antibodies bound to mouse SIRPB 1.
  • anti-SIRPB 1 antibodies were also screened for cross-reactivity to human and mouse SIRPa. In cell binding assays, none of the anti-SIRPB 1 antibodies bound cells overexpressing human or mouse SIRPa. Table 7: Cell Binding Characterization of anti-SIRPpi antibodies
  • anti-SIRP 1 antibodies were also screened for antigen specificity by using a reporter cell line expressing the luciferase gene under the control of an NFAT (nuclear factor of activated T-cells) promoter.
  • the cell line BW5147.G.1.4 (ATCC® TIB48TM), derived from mouse thymus lymphoma T lymphocytes, was infected with Cignal Lenti NFAT-luciferase virus (Qiagen), resulting inBWZ/NFAT-luciferase reporter cells.
  • cells were transduced with either a lentivirus expressing human SIRPa-DAPl2 chimera, in which the intracellular ITIM motif of SIRPa was substituted with the intracellular IT AM motif of DAP12, or with two lentiviruses expressing human SIRP 1 and human DAP12.
  • Test antibodies as well as the human IgGl isotype control, were adsorbed onto a 96-well plate at lOug/mL.
  • NFAT-luciferase reporter cells expressing the huSIRPa/DAPl2 chimera (BWZ-huSIRPa) or co-expressing huSIRP 1 and DAP12 (BWZ-huSIRP 1 ) were seeded onto plates and incubated overnight at 37C. Luciferase activity was measured by adding OneGlo Reagent (Promega) to each well and incubating the samples for 3 min at room temperature on a plate shaker. The luminescence signal was quantified using a BioTek SynergyTM Microplate Reader using GEN5TM 2.04 software. As shown in FIG.
  • the SIRP family comprises several transmembrane glycoproteins primarily expressed within the myeloid cell compartment.
  • the expression pattern of SIRPB 1 was verified on primary human cells isolated from healthy human donors. Human primary monocytes were isolated from heparinized human peripheral blood obtained from two healthy donors (Blood Centers of the Pacific) using RosetteSep Human Monocyte Enrichment Cocktail (STEMCELL Technologies), according to the manufacturer's protocol. Both SIRP 1 and TREM1 are preferentially expressed on CD 14-high monocytes.
  • Monocytes were seeded in RPMI (Invitrogen) containing 10% Fetal Calf Serum (Hy clone) and 50 ng/ml M-CSF or GM-CSF (Peprotech) to induce differentiation of M2-like or Ml -like macrophages, respectively. After 5-6 days, macrophages were harvested by scraping cells attached to plastic. Alternatively, monocytes were seeded in RPMI medium containing 10% Fetal Calf Serum (Hyclone) and 20 ng/ml IL-4 and GM-CSF (Peprotech) to induce differentiation of immature dendritic cells. After 6-7 days, dendritic cells were harvested by scraping cells attached to plastic.
  • RPMI Invitrogen
  • M-CSF M-CSF
  • GM-CSF GM-CSF
  • CD 14 expression defines a subset of primary human monocytes.
  • Classical and intermediate monocytes express high levels of CD14, whereas, non-classical monocytes lack CD14 expression.
  • SIRP 1 is abundantly expressed in CD14+ monocytes.
  • Monocytes isolated from peripheral blood as described above were cultured for 5 days with M-CSF or GM-CSF to generate M2 or Ml macrophages, respectively.
  • SIRP 1 expression decreases upon differentiation of primary monocytes into macrophages with Ml -like macrophages expressing higher levels of the receptor relative to M2-like macrophages (FIG. 4B).
  • SIRP 1 expression decreases upon differentiation of primary monocytes into immature dendritic cells. LPS-induced maturation of dendritic cells further downregulates SIRP 1 expression, in contrast to the increased expression observed for TREM1 (FIG. 4C). Unlike TREM1, LPS treatment downregulates SIRP 1 expression on DCs. Peak expression for SIRP 1 occurs in blood, spleen, and lung. Surveying SIRP 1 expression pattern from RNA-seq data sets from multiple tissue samples gathered from healthy donors confirms that SIRP 1 transcript levels predominate in the blood, consistent with high expression of the receptor in circulating monocytes and neutrophils (FIG. 5A).
  • RNA-seq data reveals that the expression pattern of SIRP 1 isoform 3 transcript differs from that of SIRP 1 isoform 1; SIRP 1 isoform 3 is mostly expressed in the brain and not peripheral tissues (FIG. 5B).
  • GTEx Genotype-Tissue Expression
  • Spleen tyrosine kinase is an intracellular signaling molecule that functions downstream of SIRPB 1 by phosphorylating several substrates, thereby facilitating the formation of a signaling complex leading to cellular activation and inflammatory processes.
  • the ability of agonist SIRPB 1 antibodies to induce Syk activation is determined by culturing mouse monocytes and measuring the phosphorylation state of Syk protein in cell extracts. In these experiments, a secondary antibody is used to cross-link anti-SIRPB 1 antibodies on cells to induce intracellular signaling.
  • BMDM Bone marrow-derived monocytes
  • WT wild-type mice
  • human SIRP 1 BAC transgenic mice from human SIRP 1 BAC transgenic mice
  • FcgR KO functional Fc receptor common gamma chain gene
  • Cell 76(3):5l9-29 are starved for 4 hours in 1% serum RPMI and then removed from tissue culture dishes with PBS-EDTA, washed with PBS, and counted.
  • the cells are coated with full-length SIRP 1 antibodies or with huIgGl isotype control for 15 minutes on ice. After washing with cold PBS, cells are incubated at 37°C for the indicated period of time in the presence of goat anti-human IgG.
  • lysis buffer 1% v/v NP-40%, 50 Mm Tns-HCl (pH 8.0), 150 mM NaCl, 1 mM EDTA, 1.5 mM MgCh, 10% glycerol, plus protease and phosphatase inhibitors
  • lysis buffer 1% v/v NP-40%, 50 Mm Tns-HCl (pH 8.0), 150 mM NaCl, 1 mM EDTA, 1.5 mM MgCh, 10% glycerol, plus protease and phosphatase inhibitors
  • immunoblots are reprobed with anti-Syk antibody (Abeam, for BMDM) or anti-Syk (Novus Biological, for human DCs). Visualization is performed with the enhanced chemiluminescence (ECL) system (GE healthcare), as described ( e.g ., Peng et al., (2010) Sci Signal., 3(122): ra38).
  • ECL enhanced chemiluminescence
  • Example 4 SIRPB 1 antibodies induce Syk phosphorylation when clustered by adjacent cells that expresses Fc gamma receptors.
  • Activation of spleen tyrosine kinase is facilitated by crosslinking two or more SIRPB 1 receptors with antibodies, thereby facilitating the formation of a signaling complex leading to cellular activation and inflammatory processes.
  • FcR Fc receptors
  • B cells accessory cells expressing Fc gamma receptors (i.e., B cells) are used to cross-link anti-SIRPB 1 antibodies to induce intracellular signaling.
  • the ability of Fc receptors to induce activation of Syk through antibody clustering is determined by culturing mouse monocytes in the presence of cells expressing Fc receptors and measuring the phosphorylation state of Syk protein in cell extracts.
  • Bone marrow-derived monocytes (BMDM) from wild-type (WT) mice and human SIRPB 1 BAC transgenic mice are starved for 4 hours in 1% serum RPMI and then removed from tissue culture dishes with PBS-EDTA, washed with PBS, and counted.
  • the cells are coated with full-length SIRPB 1 antibodies, or huIgGl isotype control for 15 minutes on ice.
  • Fc receptor expressing cells are either B cells isolated from mouse spleens using MACS microbeads (CDl9 + B-cell isolation kit Miltenyi Biotec) according to the manufacturer's protocol, or alternatively, the P815 cell line that overexpresses FcR2b and FcR3. 2x l0 6 cells/ml cells are fixed with 0.05% glutaraldehyde for 1 minute at room temperature, the reaction is stopped with ImM Glycine and cells are then washed extensively with PBS.
  • lysis buffer 1% v/v NP-40%, 50 Mm Tns-HCl (pH 8.0), 150 mM NaCl, 1 mM EDTA, 1.5 mM MgCh, 10% glycerol, plus protease and phosphatase inhibitors
  • Lysates are then immunoprecipitated with anti- Syk antibody (N-19 for BMDM or 4D10 for human DCs, Santa Cruz Biotechnology).
  • Precipitated proteins are fractionated by SDS-PAGE, transferred to PVDF membranes and probed with anti- phosphotyrosine ntibody (4G10, Millipore).
  • immunoblots are reprobed with anti-Syk antibody (Abeam, for BMDM) or anti-Syk (Novus Biological, for human DCs). Visualization is performed with the enhanced chemiluminescence (ECL) system (GE healthcare), as described ( e.g ., Peng et al, (2010) Sci Signal., 3(122): ra38).
  • ECL enhanced chemiluminescence
  • SIRPB 1 antibodies of the present disclosure was evaluated in primary human innate immune cells (e.g., monocytes and neutrophils).
  • Anti-SIRP 1 and isotype control antibodies were diluted in serum-free RPMI media and mixed with 100,000 neutrophils on 96-well plates at a concentration of 10 pg/ml.
  • Primary neutrophils were isolated with EasySepTM Direct Human Neutrophil Isolation Kit (STEMCELL) according to the manufacturer’s instructions from peripheral blood obtained the same day.
  • ROS reactive oxygen species
  • Cells were stimulated with soluble, full-length human IgGl isotype control or the anti-SIRP l antibodies SB-l, -2, -3, -4, -5, -6, -7, -8, -9, -11, -14, -15, -17, -27, -28, -31, -39, - 40, -41, -45, -46, and -49.
  • the relative fluorescence units in cells were measured at excitation wavelength 495 nm and emission wavelength 530 nm.
  • Specific fluorescence index of stimulated cells was obtained by subtraction of background fluorescence of labeled cells incubated in medium alone and/or with isotype control antibody (huIgGl). Plates were read with a BioTek SynergyTM Microplate Reader using GEN5TM 2.04 software.
  • FIG. 6A primary human neutrophils from 2 healthy donors were stimulated with 10 pg/mL of soluble full-length anti-SIRP 1 antibodies or human IgGl isotype control and labeled with 2 mM CM-H2DCFDA for 1 hour at 37°C to monitor SIRP 1 -mediated ROS production.
  • the SIRP 1 antibodies SB-l, -2, -3, and -5 proved highly agonistic in solution.
  • SIRP 1 antibodies SB-15, -17, -27, -28, -31, -39, -40, -41, -45, -46, and -49 weakly activated bIKRbI- mediated respiratory burst in solution.
  • SIRP 1 antibodies SB-l, -2, -3, -7, -8, -9, -14, 28, and -49 proved highly agonistic as plate-bound antibodies.
  • SIRP 1 antibodies SB-4, -12, -23, -26, - 33, -34, -35, -36, and -37 weakly activated SIRPpl -mediated cytokine release.
  • Example 6 SIRPB 1 increases secretion of inflammatory cytokines from macrophages.
  • BMDM bone marrow-derived macrophages
  • SIRPB 1 antibodies of the present disclosure induce changes in inflammatory cytokine production
  • primary human monocyte-derived macrophages and dendritic cells are cultured with plate-bound test antibodies in combination with non-saturating levels of TLR stimulators and the level of cytokines are measured after 24h.
  • human primary monocytes were isolated from heparinized human blood (Blood Centers of the Pacific) using RosetteSep Human Monocyte Enrichment Cocktail (STEMCELL Technologies), according to the manufacturer's protocol. Monocytes were seeded in RPMI
  • monocyte-derived macrophages and dendritic cells were stimulated overnight at 37°C with 0.5 ng/mL LPS in the presence of anti-SIRP l antibodies (SB-l, -2, -3, -4, - 5, -6, -7, -8, -9, -11, -12, -14, -15, -16, -17, -18, -19, -20, -21, -28, -32, -40, and -49) adsorbed onto 96-well plates at 10 pg/mL. The supernatant fraction was subsequently collected and assayed for TNFa release.
  • anti-SIRP l antibodies SB-l, -2, -3, -4, - 5, -6, -7, -8, -9, -11, -12, -14, -15, -16, -17, -18, -19, -20, -21, -28, -32, -40, and -49
  • LPS-induced TNFa release was decreased in dendritic cells stimulated with plate-bound anti-SIRPa antibody, SA-90, confirming that agonistic anti-SIRP 1 antibodies activate cells, whereas, agonistic SIRPa antibodies inhibit cellular activity.
  • Example 7 Anti -tumor activity of SIRPB 1 -stimulated neutrophils
  • neutrophils are efficient phagocytes for antibody- or complement-opsonized target cells, in the context of the tumor microenvironment, neutrophils generally contribute towards tumor progression, invasion, and angiogenesis.
  • tumor-associated neutrophils like other myeloid cells, retain the potential to polarize towards an anti-tumor phenotype.
  • anti-SIRP 1 antibodies were evaluated for their ability to induce neutrophil-mediated tumor cell clearance in vitro.
  • Primary neutrophils were isolated with EasySepTM Direct Human Neutrophil Isolation Kit (STEMCELL) according to the manufacturer’s instructions from peripheral blood of healthy donors obtained the same day.
  • Isolated human neutrophils were then added onto 96-well plates previously coated with 10 pg/mL anti-SIRP 1 antibodies or isotype control. Subsequently, Raji B cell lymphoma cells engineered to stably express luciferase were mixed with neutrophils in a 1 : 1 ratio with or without opsonizing antibody (anti-CD20 human IgGl). Co-cultured cells were incubated overnight at 37°C, and viable Raji cells were quantified by measuring luciferase activity following the addition of OneGlo reagent (Promega) and incubating samples at room temperature for 3 min on a plate shaker. The
  • Selection pressures used for screening the libraries included human SIRPa and SIRP 1 antigen equilibrium titration, parental antibody Fab competition kinetics, and the use of poly specificity reagent deselection (as described, for example, in WO 2014/179363; Xu et al., Protein EngDes Sel, 26(10): 663-670). FACS flow cytometry was then employed to visualize and select antibodies, using standard techniques (see, e.g., Chao et al. Nature Protocols, 2006;1 :755-768). The desired population was then carried forward into additional selection rounds. After 6 rounds of enrichment, yeast cells were plated out in order to obtain single antibody isolates, which were then produced and characterized as described in Example 1. Seventeen affinity-improved antibodies from four of the five starting parental antibodies were thus obtained.
  • Yeast clones were grown to saturation and then induced for 48 h at 30°C with shaking. After induction, yeast cells were pelleted and the supernatants were harvested for purification.
  • Immunoglobulins were purified using a Protein A column and eluted with acetic acid, pH 2.0. Fab fragments were generated by papain digestion and purified over CaptureSelect IgG-CHl affinity matrix (FifeTechnologies).
  • affinities of the anti-SIRP 1 antibodies were determined by measuring KD values by ForteBio Octet ® and Meso Scale Discovery (MSD) instrument. Octet ® affinity measurements were performed at room temperature, generally as previously described (Estep et al, MAbs. 2013 Mar- Apr;5(2):270-8). Briefly, Octet ® affinity measurements were performed by loading IgGs on-line onto AHQ sensors. Sensors were equilibrated off-line in assay buffer for 30 min and then monitored on-line for 60 seconds for baseline establishment.
  • MSD Meso Scale Discovery
  • sensors with loaded IgGs were exposed to 100 nM antigen (human SIRPa or SIRP 1 Fc fusion) for 3 min, and then transferred to assay buffer for 3 min for off-rate measurement. Additional avid binding was determined by loading biotinylated SIRP 1 monomer on SA sensors and exposuring to 100 nM IgG in solution. Monovalent binding measurements were obtained by loading human SIRP 1 Fc fusion antigens to AHQ sensor, followed by exposure to 100 nM anti-SIRP 1 antibody Fab. Additional monovalent measurements were made by loading biotinylated human SIRP 1 monomer to SA sensor followed by exposure to 100 nM Fab in solution. Kinetics data were fit using a 1 : 1 binding model in the data analysis software provided by ForteBio.
  • Affinity -matured anti-SIRPB 1 antibody clones which showed improved affinity compared to the respective parental antibody, were characterized further. After initial screening of all affinity- matured antibody clones, clones for each parental antibody were selected for further analysis. Antibody heavy chain and light chain variable domain sequences
  • the amino acid sequences encoding the light chain variable and the heavy chain variable domains of the affinity matured antibodies were determined.
  • the Kabat light chain HVR sequences of the affinity matured antibodies are set forth in Table 8.
  • the Kabat heavy chain HVR sequences of the antibodies are set forth in Table 9.
  • the Kabat heavy chain framework (FR) sequences of the antibodies are set forth in Table 10.
  • the Kabat light chain framework (FR) sequences of the antibodies are set forth in Table 11.
  • Table 8 Kabat light chain HVR sequences of affinity matured anti-SIRPpi antibodies
  • a final set of affinity matured anti-SIRPB 1 antibodies were selected based on antigen binding affinities. Antibodies that were positive for binding to human SIRPB 1 were tested for cross reactivity to human SIRPa. The biochemical characteristics of each antibody are listed below in Table 12. In Table 12,“N.B.” refers to antibodies for which there is no binding to the indicated antigen;“P.F.” refers to antibodies for which antigen binding kinetics show poor fit to 1 : 1 binding model;“N.M.” refers to not measurable.
  • Table 13 shows the mean fluorescence intensity (MFI) values of affinity matured anti- SIRP 1 antibodies binding to the Chinese hamster ovary (CHO) cell line expressing low levels of human SIRP 1.
  • the human IgGl isotype control established the background fluorescence signal set to 1.
  • the anti-SIRP 1 antibodies were also screened for surface binding to CHO cells overexpressing mouse SIRP 1.
  • none of the test antibodies showed significant binding to mouse SIRP 1 , as clones were originally selected for binding the human antigen.
  • anti- SIRP 1 antibodies were also screened for cross-reactivity to human SIRPa. In cell binding assays, none of the anti-SIRP 1 antibodies bound cells overexpressing human SIRPa.
  • Example 9 Affinity matured anti-SIRPB 1 antibodies increase secretion of inflammatory cytokines from dendritic cells.
  • Dendritic cells were harvested by scraping cells attached to plastic. Dendritic cells are plated on 96-well plates coated with indicated antibody at 10 5 cells/well and incubated for 24h at 37°C. Cells are co-stimulated with TLR4 agonist, LPS ⁇ Salmonella abortus equi).
  • FIG. 9 monocyte-derived dendritic cells from 3 healthy donors were stimulated overnight at 37°C with 0.5 ng/mL LPS in the presence of indicated affinity matured anti-SIRP 1 antibodies adsorbed onto 96-well plates at 2 pg/mL. The supernatant fraction was subsequently collected and assayed for TNFa release.
  • the SIRP l antibodies SB-l-3, SB-2-8, and SB-8-13 proved highly agonistic as plate-bound antibodies.
  • remaining SIRP 1 antibodies, SB-2-8, SB-8-15, and SB-40-20 weakly activated SIRPpl -mediated cytokine release.
  • isoform 1 is the full-length version of the receptor primarily expressed in the periphery.
  • Recombinant antigen based on the sequence of SIRPB 1 isoform 1 was used for selecting anti-SIRPB 1 antibodies from yeast library pools.
  • recombinant antigens based on human SIRPB 1 isoform 3 sequence and cynomolgus SIRPB 1 isoform 1 sequence were produced by transient transfection in HEK293 cells.
  • FIG. 10A-10B shows binding curves of anti-SIRP 1 antibodies bound to indicated SIRP 1 antigens based on ELISA data. Eight anti-SIRP 1 antibodies (four parental antibodies and four corresponding affinity matured antibodies) were selected for analysis. EC50 values calculated from these binding curves demonstrated that all eight anti-SIRP 1 antibodies recognized human SIRP 1 isoform 1 with high affinity. For example, the EC50 values for SB-l and SB-l-2 are 0.034 nM and 0.045 nM, respectively. The EC50 values for SB-2 and SB-2-7 are 0.032 nM and 0.029 nM, respectively.
  • the EC50 values for SB-8 and SB-8-13 are 0.022 nM and 0.019 nM, respectively.
  • the EC50 values for SB-40 and SB-40-21 are 0.040 nM and 0.025 nM, respectively.
  • the anti- SIRP 1 antibodies tested did not cross-react to either human SIRP 1 isoform 3 or to cynomolgus SIRP 1.
  • Example 11 Analysis of the effect of anti-SIRPB 1 antibodies in increasing recruitment of immune cells in vivo
  • mice receive first an IP injection of inflammatory cells (neutrophil granulocytes, monocytes, and macrophages) in the peritoneal cavity (PEC) of human SIRP 1 BAC-transgenic mice after intraperitoneal (IP) administration of either antibody alone or in combination with LPS is evaluated as follows. Briefly, mice receive first an IP injection of inflammatory cells (neutrophil granulocytes, monocytes, and macrophages) in the peritoneal cavity (PEC) of human SIRP 1 BAC-transgenic mice after intraperitoneal (IP) administration of either antibody alone or in combination with LPS is evaluated as follows. Briefly, mice receive first an IP injection of inflammatory cells (neutrophil granulocytes, monocytes, and macrophages) in the peritoneal cavity (PEC) of human SIRP 1 BAC-transgenic mice after intraperitoneal (IP) administration of either antibody alone or in combination with LPS is evaluated as follows. Briefly, mice receive first an IP injection of
  • anti-SIRPB 1 antibody or isotype control antibody mlgGl (clone MOPC-21, Bioxcell).
  • mice receive an IP injection of 4 mg/kg LPS, or PBS as a control.
  • cells are harvested from the PEC as described (see, e.g., Gawish R el al, 2014 FASEB J) and analyzed by FACS.
  • FACS analysis PEC cells are incubated with anti-CDl lb- Pacific Blue, anti-CDl lc PeCy7, anti-MCH-II- APCCy7, anti-Grl-FITC, anti-Ly6G-PE and a viability die (Life Technologies, Cat# L34957) for 1 hour on ice, then washed twice with cold FACS buffer. 4% PFA-fixed samples are then acquired. Data are acquired on a BD FACS CANTO II cytometer (Becton Dickinson) and analyzed with FlowJo software.
  • mice Groups of 3 human SIRPB 1 BAC-transgenic mice (females, 8 weeks old) are challenged subcutaneously with lxl 0 6 MC38 or CT26 colon carcinoma cells, or EMT-6 murine mammary carcinoma cells, suspended in IOOmI PBS. Animals are anesthetized with isoflurane prior to implant. When the tumors reach a size of 700-1000 mm 3 , tumors are explanted to analyze SIRPB 1 expression in the tumor microenvironment by FACS. As a comparison, the spleen of the tumor bearing mice or control spleen of naive mice is also analyzed.
  • tumor and spleens are incubated in PBS containing 1 mg/ml collagenase and then processed through a cell strained to obtain a single cell suspension.
  • Cells are then incubated with anti-CD45-PerCp-Cy7, anti-CDl lb- PerCP-Cy5.5, anti- CD3-PC, anti-Grl-FITC, anti-NKl.l-PE, anti-SIRP 1 -APC antibodies and a viability die (Life Technologies, Cat# L34957) for 30min on ice, then washed twice with cold FACS buffer. 4% PFA-fixed samples are then acquired. Data are acquired on a BD FACS CANTO II cytometer (Becton Dickinson) and analyzed with FlowJo software. These studies provide support that myeloid cells or cells of myeloid lineage express SIRP 1 in the tumor environment.
  • Example 14 Anti-SIRPB 1 antibodies induce the expression of CD83 and CD86 on human dendritic cells (PCs ' )
  • both plate-bound and soluble antibodies are incubated with dendritic cells (DCs), and the expression of CD83, CD86, CCR7, and phosphorylated ERK are measured.
  • DCs dendritic cells
  • Antibodies are plated overnight at 4°C in 12 well plates at 2 or 10 pg/ml in PBS. Wells are washed 3X with PBS the next day.
  • Primary human monocytes isolated from peripheral blood of healthy donors are added to antibody coated wells in RPMI media supplemented with 10% FBS and 20 ng/mL IL-4 and GM-CSF and incubated at 37°C, 5% CCh for 5 days.
  • CDla+/HLA-/DR+ cell populations For intracellular ERK phosphorylation, cells are fixed with 1% formaldehyde, permeabilized with cytofix/cytoperm kit (BD), and intracellular Erk phopshorylation is determined with flow cytometry after staining with PE -ERK antibody (BD).
  • BD cytofix/cytoperm kit
  • Erk phopshorylation is determined with flow cytometry after staining with PE -ERK antibody (BD).
  • Example 15 Screening for anti-SIRPB 1 and/or anti-SIRPB 1 bispecific antibodies that induce phosphorylation of SIRPBE DAP 12. SYK. ERK. and AKT. which indicate activation of the PI3K pathway
  • RIP A radioimmunoprecipitation assay
  • anti-SIRPB 1 antibodies of the present invention induce phosphorylation of SIRPB 1 , DAP12, SYK, ERK, and AKT. Additionally, these studies provide support that anti-SIRPB 1 antibodies of the present invention are effective at activating the PI3K pathway.
  • SIRPB 1 and TREM2 are DAPl2-associated receptors expressed on myeloid cells.
  • SIRPB 1 modifies the expression of TREM2
  • macrophages were cultured on plate-bound antibodies and analyzed for cell surface levels of TREM2 by flow cytometry.
  • Antibodies (SB-l-3, SB-2-8, SB-8-13, SB-8-15, SB-40-20, and huIgGl control) were adsorbed onto 96-well plates at 37°C for 4 hours at 2 pg/mL in PBS. Wells were washed twice with PBS. Monocyte-derived human macrophages, differentiating in culture with M-CSF for 5-6 days, were harvested and plated at 100,000 cells per well. Macrophages were incubated overnight at 37°C, 5% CO2. FACS analysis of TREM2 expression was performed on a BD FACS Canto II and data analysis was performed with FlowJo (TreeStar) software. [0291] As shown in FIG.
  • Plate-bound anti-SIRPpl antibodies SB-2-8, SB-8-15, and SB-40- 20 increased TREM2 expression approximately 2-fold relative to huIgGl isotype control treated macrophages obtained from 3 healthy donors. In 2 out of 3 healthy donors (624 and 626), however, plate-bound anti-SIRP 1 antibodies SB- 1-3 and SB-8- 13 only partially increased TREM2 expression relative to huIgGl isotype control treated macrophages. In macrophages from a third healthy donor (625), plate-bound SB- 1-3 and SB-8- 13 increased TREM2 expression approximately 2-fold relative to huIgGl isotype control. Based on these results, anti-SIRP 1 antibodies SB-2-8, SB-8-15, and SB-40-20 function as agonists to induce cell surface TREM2 expression.
  • Antibodies (SB-l-3, SB-2-8, SB-8-13, SB-8-15, SB-40-20, and huIgGl control) were adsorbed onto 96-well plates at 37°C for 4 hours at 2 pg/mL in PBS. Wells were washed twice with PBS. Monocyte-derived human macrophages, differentiating in culture with M-CSF for 5-6 days, were harvested and washed to remove residual M-CSF. Macrophages were resuspended in RPMI growth media supplemented with 10% FBS and 1% Penn/Strep at 500,000 cell per mL. Cells were diluted with equal volume of PBS and 100 pL containing 25,000 cells were added to each well.
  • Macrophages were incubated at 37°C for 2 days. Analysis of viability was performed using Cell Titer Glo kit (Promega), a reagent that produces a luminescence signal relative to ATP concentration in the sample. Plates were read with a Biotek Synergy Microplate Reader using GEN5 2.04 software.
  • Anti-SIRPB 1 antibodies SB-l-3 and SB-2-8 or the human IgGl isotype control were coated onto 96- well plates at 37°C for 4 hours starting at 10 pg/mL in PBS and serially diluted 3-fold.
  • macrophages were diluted with equal volume of PBS and 100 pL containing 25,000 cells were added to each well. Macrophages were incubated at 37°C for 2 days. Analysis of viability was performed using Cell Titer Glo kit (Promega) and plates were read with a Biotek Synergy
  • plate-bound, full-length anti-SIRPB 1 antibody SB- 1-3 increased macrophage viability in a dose-dependent manner relative to isotype control treated cells.
  • plate-bound, full-length anti-SIRPB 1 antibody SB-2-8 failed to increase macrophage viability even at high concentrations of coated protein.
  • anti-SIRPB 1 antibodies SB- 1-3, SB-8- 13, and SB-8- 15 demonstrated agonistic activity in this assay.
  • Example 18 Additive and synergistic effect of combination treatment of macrophages with anti- SIRPB 1 and anti-TREM2 antibodies
  • Agonistic anti-TREM2 antibodies have been shown to increase macrophage viability when added to cells in a soluble format. Viability assays were performed to determine if co-stimulating macrophages with anti-SIRPB 1 antibodies further enhances the agonist activity anti-TREM2 antibody.
  • Anti-SIRPB 1 antibodies SB-l-3 and SB-2-8 or the human IgGl isotype control were coated onto 96-well plates at 37°C for 4 hours at 10 pg/mL in PBS.
  • macrophages were diluted with equal volume of PBS and 100 pL containing 25,000 cells were added to each well. Where indicated, macrophages were also treated with 50 pg/mL of anti-TREM2 antibody. Macrophages were incubated at 37°C for 2 days. Analysis of viability was performed using Cell Titer Glo kit (Promega) and plates were read with a Biotek Synergy Microplate Reader using GEN5 2.04 software.
  • adding agonist anti-TREM2 antibody to cells cultured on human IgGl isotype control significantly increased macrophage viability relative to cells cultured in the absence of agonist anti-TREM2 antibody, establishing the reproducibility of this assay.
  • Human SIRPB 1 BAC transgenic mice that recapitulate the expression of the human antigen in mouse myeloid cells were generated.
  • Murine bone marrow cells from human SIRPB 1 transgenic mice were obtained by flushing tibia and femurs with PBS. Bone marrow cells were cultured in RPMI media supplemented with 50 ng/mL M-CSF to differentiate macrophages or 10 ng/mL GM- CSF to differentiate dendritic cells.
  • Anti-SIRP 1 antibodies were evaluated for their ability to increase murine myeloid cell viability.
  • Anti-SIRP l antibodies SB- 1-3, SB-2-8, and SB-8- 13 or the human IgGl isotype control were coated onto 96-well plates at 37°C for 4 hours at 10 pg/mL in PBS. Macrophages and dendritic cells were diluted with equal volume of PBS and 100 pL containing 25,000 cells were added to each well. Cells were incubated at 37°C for 2 days. Analysis of viability was performed using Cell Titer Glo kit (Promega) and plates were read with a Biotek Synergy Microplate Reader using GEN5 2.04 software. As shown in FIG.
  • anti-SIRP 1 antibodies SB- 1-3 and SB-8- 13 enhanced macrophage viability relative to isotype control treated cells.
  • anti-SIRP 1 antibody SB-2-8 failed to increase macrophage viability.
  • FIG. 14B when dendritic cells (DC) were cultured on plate-bound antibodies, all anti-SIRP 1 antibodies increased DC viability relative to isotype control treated cells. However, only anti-SIRP 1 antibody SB-8- 13 treated DC showed increased viability relative to untreated dendritic cells (No Ab.). Thus, anti- SIRP 1 antibodies show agonistic activity with murine myeloid cells expressing human SIRP 1.
  • SIRP 1 A characteristic feature of proteins of the SIRP family is their extensive amino acid sequence conservation in the extracellular domain. For example, the extracellular region of SIRP 1 shares -90% sequence identity with SIRPa and -77% sequence identity with SIRPy. Antibodies developed against one SIRP protein family member often cross-react with multiple SIRP receptor family members.
  • SIRPa contains a cytoplasmic ITIM motif to mediate immune suppression upon binding to CD47.
  • SIRP 1 which lacks intracellular signaling motifs and does not bind CD47, associates with the ITAM-containing DAP12 adaptor protein to propagate activation signals.
  • SIRPy unlike other SIRP receptors that are primarily expressed on myeloid cells, is expressed on lymphocytes and NK cells and serves to stabilize cell-cell adhesion during antigen presentation to T cells.
  • BW5147.G.1.4 cells an immortalized mouse T-cell line, were engineered to overexpress human SIRPB 1 (BWZ-huSIRPB 1 ) or human SIRPa (BWZ-huSIRPa).
  • Jurkat cells an immortalized human T cell line, endogenously express SIRPy.
  • antibody 18D5 an anti-SIRP antibody that cross-reacts with SIRPa and SIRPP
  • antibody KWAR23 an anti-SIRP antibody that cross-reacts with SIRPa, SIRPP, and SIRPy
  • These antibodies were recombinantly produced on a human IgG4 backbone.
  • a commercially available anti-SIRPy antibody (clone LSB2.20; Biolegend, San Diego) conjugated with phycoerythrin (PE) fluorophore was used to verify SIRPy expression on Jurkat cells.
  • PE phycoerythrin
  • Fig. 15A anti-SIRP 1 antibodies bound BWZ-huSIRP 1 cells with distinct EC50 profiles.
  • Anti- Si RP 1 antibody SB 1-3 showed the best apparent affinity relative to other anti-SIRP 1 antibodies, as well as compared to that of the positive control antibody, 18D5.
  • Anti-SIRP 1 antibodies failed to bind cells expressing SIRPa (Fig. 15B) or SIRPy (Fig. 15C) demonstrating that these antibodies are antigen-specific.
  • Table 14 EC50 values of antibodies binding BWZ-human SIRPpi cells
  • SB-28 Heavy Chain Variable Region QLQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIGSISYSGSTYYNPSLKS RVTISVDTSKNQFSLKLSSVTAADTAVYYCARDPRDYSSGSSGGGWGYFDLWGRGTLVTVSS (SEQ ID NO: 321)

Abstract

La présente invention concerne d'une manière générale des compositions qui comprennent des anticorps, par exemple, des anticorps monoclonaux, des fragments d'anticorps, etc., qui se lient de manière spécifique à un polypeptide SIRPβ1, par exemple, SIRPβ1 humain, et l'utilisation desdites compositions dans la prévention, la réduction des risques, ou le traitement d'un individu en ayant besoin.
PCT/US2019/039757 2018-06-29 2019-06-28 Anticorps anti-sirp-bêta1 et procédés d'utilisation associés WO2020006374A2 (fr)

Priority Applications (12)

Application Number Priority Date Filing Date Title
JP2020572914A JP2021529520A (ja) 2018-06-29 2019-06-28 抗sirp−ベータ1抗体及びその使用方法
KR1020217002521A KR20210025614A (ko) 2018-06-29 2019-06-28 항-sirp-베타1 항체 및 그의 사용 방법
CN201980040821.9A CN112384532A (zh) 2018-06-29 2019-06-28 抗SIRP-β1抗体及其使用方法
CA3099176A CA3099176A1 (fr) 2018-06-29 2019-06-28 Anticorps anti-sirp-beta1 et procedes d'utilisation associes
US17/256,508 US20210277113A1 (en) 2018-06-29 2019-06-28 Anti-SIRP-Beta1 Antibodies and Methods of Use Thereof
EP19744960.6A EP3814377A2 (fr) 2018-06-29 2019-06-28 Anticorps anti-sirp-bêta1 et procédés d'utilisation associés
MX2020013324A MX2020013324A (es) 2018-06-29 2019-06-28 Anticuerpos anti proteína reguladora de señales beta 1 (sirp-beta1) y métodos de uso de los mismos.
AU2019293589A AU2019293589A1 (en) 2018-06-29 2019-06-28 Anti-SIRP-beta1 antibodies and methods of use thereof
EA202190138A EA202190138A1 (ru) 2018-06-29 2019-06-28 Анти-sirp-бета1 антитела и способы их использования
SG11202010990TA SG11202010990TA (en) 2018-06-29 2019-06-28 Anti-sirp-beta1 antibodies and methods of use thereof
BR112020026819-4A BR112020026819A2 (pt) 2018-06-29 2019-06-28 anticorpos isolados, ácido nucleico, vetor, células hospedeiras, método de produção de um anticorpo, composição farmacêutica, métodos para tratar o câncer e para tratar uma doença e usos de um anticorpo
IL279648A IL279648A (en) 2018-06-29 2020-12-21 Anti-SIRP-in-cell 1 antibodies and methods of using them

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862691913P 2018-06-29 2018-06-29
US62/691,913 2018-06-29

Publications (2)

Publication Number Publication Date
WO2020006374A2 true WO2020006374A2 (fr) 2020-01-02
WO2020006374A3 WO2020006374A3 (fr) 2020-02-20

Family

ID=67439371

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/039757 WO2020006374A2 (fr) 2018-06-29 2019-06-28 Anticorps anti-sirp-bêta1 et procédés d'utilisation associés

Country Status (14)

Country Link
US (1) US20210277113A1 (fr)
EP (1) EP3814377A2 (fr)
JP (1) JP2021529520A (fr)
KR (1) KR20210025614A (fr)
CN (1) CN112384532A (fr)
AU (1) AU2019293589A1 (fr)
BR (1) BR112020026819A2 (fr)
CA (1) CA3099176A1 (fr)
EA (1) EA202190138A1 (fr)
IL (1) IL279648A (fr)
MA (1) MA53015A (fr)
MX (1) MX2020013324A (fr)
SG (1) SG11202010990TA (fr)
WO (1) WO2020006374A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021217024A1 (fr) * 2020-04-24 2021-10-28 Millennium Pharmaceuticals, Inc. Anticorps anti-cd19 et leurs utilisations
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
WO2022115350A1 (fr) * 2020-11-30 2022-06-02 Merck Sharp & Dohme Corp. Liants d'arginase 1 pour inhiber l'activité de l'arginase 1
WO2022120064A1 (fr) * 2020-12-03 2022-06-09 The Board Of Regents Of The University Of Texas System Méthodes d'identification d'anticorps bloquant lilrb
US11939367B2 (en) 2015-06-30 2024-03-26 Sanford Burnham Prebys Medical Discovery Institute BTLA fusion protein agonists and uses thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114478802B (zh) * 2022-01-28 2023-05-26 郑州大学 一种嵌合抗原受体及其应用

Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4301144A (en) 1979-07-11 1981-11-17 Ajinomoto Company, Incorporated Blood substitute containing modified hemoglobin
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4670417A (en) 1985-06-19 1987-06-02 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1993001161A1 (fr) 1991-07-11 1993-01-21 Pfizer Limited Procede de preparation d'intermediaires de sertraline
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
EP0546073A1 (fr) 1990-08-29 1993-06-16 Genpharm Int Animaux non humains transgeniques capables de produire des anticorps heterologues.
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1997011971A1 (fr) 1995-09-28 1997-04-03 Alexion Pharmaceuticals, Inc. Proteines d'interaction de cellules porcines
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999058572A1 (fr) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Molecules de liaison derivees d'immunoglobulines ne declenchant pas de lyse dependante du complement
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6133426A (en) 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
US20070148167A1 (en) 2005-02-14 2007-06-28 Strohl William R Non-immunostimulatory antibody and compositions containing the same
WO2007106585A1 (fr) 2006-03-15 2007-09-20 Alexion Pharmaceuticals, Inc. Traitement de patients souffrant d'hemoglobinurie paroxystique nocturne par un inhibiteur de complement
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008079246A2 (fr) 2006-12-21 2008-07-03 Medarex, Inc. Anticorps anti-cd44
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
WO2009036379A2 (fr) 2007-09-14 2009-03-19 Adimab, Inc. Bibliothèques d'anticorps synthétiques rationnelles et leurs utilisations
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2010105256A1 (fr) 2009-03-13 2010-09-16 Adimab, Inc. Banques d'anticorps synthétiques, conçues de façon rationnelle, et leurs utilisations
WO2012009568A2 (fr) 2010-07-16 2012-01-19 Adimab, Llc Banques d'anticorps
WO2013026833A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t.
US20140242095A1 (en) 2011-10-19 2014-08-28 University Health Network Antibodies and antibody fragments targeting sirp-alpha and their use in treating hematologic cancers
WO2014179363A1 (fr) 2013-04-29 2014-11-06 Adimab, Llc Réactifs multispécificité, procédés de préparation et d'utilisation associés

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4224586B2 (ja) * 2004-04-28 2009-02-18 国立大学法人群馬大学 マクロファージ活性化剤並びにその製造方法及びスクリーニング方法
MY146381A (en) * 2004-12-22 2012-08-15 Amgen Inc Compositions and methods relating relating to anti-igf-1 receptor antibodies
CN101880324B (zh) * 2010-05-25 2012-10-17 中国人民解放军第二军医大学 一种抗人SIRPα的单克隆抗体及其细胞株、制备方法和应用
EP3553086A1 (fr) * 2012-05-31 2019-10-16 Sorrento Therapeutics Inc. Protéines de liaison à un antigène se liant à pd-l1
AU2015229448B2 (en) * 2014-03-11 2020-09-03 The Board Of Trustees Of The Leland Stanford Junior University Anti SIRP-alpha antibodies and Bi-specific Macrophage Enhancing antibodies
GB201413357D0 (en) * 2014-07-28 2014-09-10 Philogen Spa Antibodies for treatment and diagnosis
TW201632559A (zh) * 2015-02-22 2016-09-16 索倫多醫療公司 結合cd137之抗體治療劑
CA2996059A1 (fr) * 2015-08-28 2017-03-09 Alector Llc Anticorps anti-siglec-7 et leurs methodes d'utilisation
US10358497B2 (en) * 2015-09-29 2019-07-23 Amgen Inc. Methods of treating cardiovascular disease with an ASGR inhibitor
US11274159B2 (en) * 2017-05-16 2022-03-15 Byondis B.V. Anti-SIRPα antibodies

Patent Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4301144A (en) 1979-07-11 1981-11-17 Ajinomoto Company, Incorporated Blood substitute containing modified hemoglobin
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4670417A (en) 1985-06-19 1987-06-02 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0546073A1 (fr) 1990-08-29 1993-06-16 Genpharm Int Animaux non humains transgeniques capables de produire des anticorps heterologues.
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (fr) 1991-07-11 1993-01-21 Pfizer Limited Procede de preparation d'intermediaires de sertraline
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5869619A (en) 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
WO1993016185A2 (fr) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Proteine de liaison biosynthetique pour marqueur de cancer
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
WO1997011971A1 (fr) 1995-09-28 1997-04-03 Alexion Pharmaceuticals, Inc. Proteines d'interaction de cellules porcines
US6133426A (en) 1997-02-21 2000-10-17 Genentech, Inc. Humanized anti-IL-8 monoclonal antibodies
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO1999058572A1 (fr) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Molecules de liaison derivees d'immunoglobulines ne declenchant pas de lyse dependante du complement
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
US20070148167A1 (en) 2005-02-14 2007-06-28 Strohl William R Non-immunostimulatory antibody and compositions containing the same
WO2007106585A1 (fr) 2006-03-15 2007-09-20 Alexion Pharmaceuticals, Inc. Traitement de patients souffrant d'hemoglobinurie paroxystique nocturne par un inhibiteur de complement
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008079246A2 (fr) 2006-12-21 2008-07-03 Medarex, Inc. Anticorps anti-cd44
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
WO2009036379A2 (fr) 2007-09-14 2009-03-19 Adimab, Inc. Bibliothèques d'anticorps synthétiques rationnelles et leurs utilisations
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2010105256A1 (fr) 2009-03-13 2010-09-16 Adimab, Inc. Banques d'anticorps synthétiques, conçues de façon rationnelle, et leurs utilisations
WO2012009568A2 (fr) 2010-07-16 2012-01-19 Adimab, Llc Banques d'anticorps
WO2013026833A1 (fr) 2011-08-23 2013-02-28 Roche Glycart Ag Molécules bispécifiques de liaison à l'antigène activant les lymphocytes t.
US20140242095A1 (en) 2011-10-19 2014-08-28 University Health Network Antibodies and antibody fragments targeting sirp-alpha and their use in treating hematologic cancers
WO2014179363A1 (fr) 2013-04-29 2014-11-06 Adimab, Llc Réactifs multispécificité, procédés de préparation et d'utilisation associés

Non-Patent Citations (103)

* Cited by examiner, † Cited by third party
Title
"Uniprot", Database accession no. Q8BFX8
ALEGRE ET AL., TRANSPLANTATION, vol. 57, 1994, pages 1537 - 1543. 31
ALMAGROFRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 161 9 - 1633
ANGAL ET AL., MOL IMMUNOL, vol. 30, 1993, pages 105 - 108
ARMOUR ET AL., EUR JIMMUNOL, vol. 29, 1999, pages 2613 - 2624
ARMOUR ET AL., IMMUNOLOGY, vol. 40, 2003, pages 585 - 593
ARMOUR ET AL., MOLECULAR IMMUNOLOGY, vol. 40, 2003, pages 585 - 593
ARMOUR ET AL., THE HAEMATOLOGY JOURNAL, vol. l, no. l, 2000, pages 27
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
BARBAS ET AL., PROC NAT. ACAD. SCI. USA, vol. 91, 1994, pages 3809 - 3813
BARTHOLOMAEUS P J, IMMUNOL, vol. 192, 2014, pages 2091 - 2098
BEATTY ET AL., NEURON, vol. 36, 2002, pages 375 - 386
BOERNER ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
BOLT S ET AL., EUR JIMMUNOL, vol. 23, 1993, pages 403 - 411
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285
CHAO ET AL., NATURE PROTOCOLS, vol. 1, 2006, pages 755 - 768
CHEN ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
CHEUNG ET AL., VIROLOGY, vol. 176, 1990, pages 546 - 552
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHU ET AL., MOL IMMUNOL, vol. 45, 2008, pages 3926 - 3933
COLE ET AL., TRANSPLANTATION, vol. 68, 1999, pages 563 - 571
CRUTS, MVAN BROECKHOVEN, C., TRENDS GENET, vol. 24, 2008, pages 186 - 194
DANEMAN ET AL., PLOSONE, vol. 5, no. 10, 2010, pages el3741
DIETRICH ET AL., J IMMUNOL, vol. 164, 2000, pages 1925 - 1933
DUCRY ET AL., BIOCONJUGATE CHEMISTRY, vol. 21, no. 1, 2010, pages 5 - 13
ESTEP ET AL., MABS, vol. 5, no. 2, March 2013 (2013-03-01), pages 270 - 8
EVANS ET AL., J. MED. CHEM., vol. 30, 1987, pages 1229
FAUCHERE, J. ADV. DRUG RES., vol. 15, 1986, pages 29
FELLOUSE, PROC. NATL. ACAD. SCI. USA, vol. 101, no. 34, 2004, pages 12467 - 12472
GABATHULER R, NEUROBIOL. DIS., vol. 37, 2010, pages 48 - 57
GAIKWAD, AM J PATHOL, vol. 175, 2009, pages 2528 - 2539
GAWISH R ET AL., FASEB J, 2014
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GIEHL ET AL., PROC. NATL. ACAD. SCI USA, vol. 101, 2004, pages 6226 - 30
GRAHAM ET AL., J. GEN VIROL., vol. 36, 1977, pages 59
GRIFFITHS ET AL., EMBO J., vol. 12, 1993, pages 725 - 734
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOOGENBOOM ET AL., J. MOL. BIOL., vol. 227, 1992, pages 381 - 388
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
HUTCHINS ET AL., PROC NATLACAD SCI USA, vol. 92, 1995, pages 11980 - 11984
HUTTON, M. ET AL., NATURE, vol. 393, 1998, pages 702 - 705
JACKSON ET AL., J. IMMUNOL., vol. 154, no. 7, 1995, pages 3310 - 2004
JANE DE LARTIGUE, ONCLIVE, 5 July 2012 (2012-07-05)
KANDA ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 680 - 688
KIRKLAND ET AL., J. IMMUNOL., vol. 137, 1986, pages 3614 - 3619
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 3001
KYTE ET AL., J. MOL. BIOL., vol. 157, 1982, pages 105 - 131
LAIRD, AS ET AL., PLOS ONE, vol. 5, 2010, pages el3368
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LAZAR ET AL., PROC NATL ACAD SCI USA, vol. 103, 2006, pages 4005 - 4010
LEE ET AL., J. IMMUNOL. METHODS, vol. 284, no. 2, 2004, pages 119 - 132
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 1, no. 03, 2006, pages 3557 - 3562
LIGHTLE ET AL., PROTEIN SCI., vol. 19, 2010, pages 753 - 762
LIU ET AL., J BIOL CHEM, vol. 280, 2005, pages 36132 - 36140
LONBERG, CURR. OPIN. IMMUNOL., vol. 20, 2008, pages 450 - 459
LYNAUGH ET AL., MABS, vol. 5, no. 5, October 2013 (2013-10-01), pages 641 - 45
MARKS ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 779 - 783
MATHER ET AL., ANNALS N.Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68
MATHER, BIOL. REPROD., vol. 23, 1980, pages 243 - 251
MCEARCHERN ET AL., BLOOD, vol. 109, 2007, pages 1185 - 1192
MILSTEINCUELLO, NATURE, vol. 305, 1983, pages 537
MOLDENHAUER ET AL., SCAND. J. IMMUNOL., vol. 32, 1990, pages 77 - 82
MOREL ET AL., MOLEC. IMMUNOL., vol. 25, 1988, pages 7 - 15
NEARY, D. ET AL., NEUROLOGY, vol. 51, 1998, pages 1546 - 1554
NEUMANN, M. ET AL., ARCH. NEUROL., vol. 64, 2007, pages 1388 - 1394
OKAZAKI ET AL., J. MOL. BIOL., vol. 336, no. 5, 2004, pages 1239 - 1249
PENG ET AL., SCI SIGNAL, vol. 3, no. 122, 2010, pages ra38
PENG ET AL., SCI SIGNAL., vol. 3, no. 122, 2010, pages ra38
PETERS ET AL., JBIOL CHEM, vol. 287, no. 29, 2012, pages 24525 - 33
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
RATNAVALLI, E.BRAYNE, C.DAWSON, KHODGES, J. R., NEUROLOGY, vol. 58, 2002, pages 1615 - 1621
REDDY ET AL., J. IMMUNOLOGY, vol. 164, 2000, pages 1925 - 1933
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
RIZOGIERASCH, ANN. REV. BIOCHEM., vol. 61, 1992, pages 387
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
SAZINSKY ET AL., PROC NATL ACAD SCI USA 2008, vol. 105, 2008, pages 20167 - 20172
SAZINSKY ET AL., PROC NATL ACAD SCI USA, vol. 105, 2008, pages 20167 - 20172
SCHIER ET AL., GENE, vol. 169, 1995, pages 147 - 155
SCHYMICK, JC ET AL., J NEUROL NEUROSURG PSYCHIATRY, vol. 78, 2007, pages 754 - 6
SHIELDS ET AL., R. J. BIOL. CHEM., vol. 276, 2001, pages 6591 - 6604
SIEGEL ET AL., J IMMUNOL METHODS, vol. 286, no. l-2, March 2004 (2004-03-01), pages 141 - 53
STAHLI ET AL., METHODS IN ENZYMOLOGY, vol. 9, 1983, pages 242 - 253
TAKAI T, CELL, vol. 76, no. 3, 1994, pages 519 - 29
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216
VAN DIJK ET AL., CURR. OPIN. PHARMACOL., vol. 5, 2001, pages 368 - 74
VOLLMERS ET AL., HISTOLOGY AND HISTOPATHOLOGY, vol. 20, no. 3, 2005, pages 927 - 937
VOLLMERS ET AL., METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 27, no. 3, 2005, pages 185 - 91
WHITE AL, CANCER IMMUNOL IMMUNOTHER, vol. 62, 2013, pages 941 - 948
WHITE ET AL., CANCER CELL, vol. 27, 2015, pages 138 - 148
WILSON ET AL., CANCER CELL, vol. 19, 2011, pages 101 - 113
WINTER ET AL., ANN. REV. IMMUNOL., vol. 12, 1994, pages 433 - 455
XU ET AL., CELL IMMUNOL, vol. 200, 2000, pages 16 - 26
XU ET AL., PEDS, vol. 26, no. 10, October 2013 (2013-10-01), pages 663 - 70
XU ET AL., PROTEIN ENG DES SEL, vol. 26, no. 10, October 2013 (2013-10-01), pages 663 - 70
XU ET AL., PROTEIN ENG DES SEL, vol. 26, no. 10, pages 663 - 670
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG, vol. 87, 2004, pages 614
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614
YAZAKIWU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268
ZAPATA ET AL., PROTEIN ENG, vol. 8, no. 10, 1995, pages 1057 - 1062
ZHU Y, CANCER RES., vol. 74, no. 18, 15 September 2014 (2014-09-15), pages 5057 - 69

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11939367B2 (en) 2015-06-30 2024-03-26 Sanford Burnham Prebys Medical Discovery Institute BTLA fusion protein agonists and uses thereof
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
WO2021217024A1 (fr) * 2020-04-24 2021-10-28 Millennium Pharmaceuticals, Inc. Anticorps anti-cd19 et leurs utilisations
WO2022115350A1 (fr) * 2020-11-30 2022-06-02 Merck Sharp & Dohme Corp. Liants d'arginase 1 pour inhiber l'activité de l'arginase 1
WO2022120064A1 (fr) * 2020-12-03 2022-06-09 The Board Of Regents Of The University Of Texas System Méthodes d'identification d'anticorps bloquant lilrb

Also Published As

Publication number Publication date
IL279648A (en) 2021-03-01
CA3099176A1 (fr) 2020-01-02
AU2019293589A1 (en) 2021-01-21
EP3814377A2 (fr) 2021-05-05
BR112020026819A2 (pt) 2021-04-20
SG11202010990TA (en) 2020-12-30
MA53015A (fr) 2021-05-05
MX2020013324A (es) 2021-05-12
CN112384532A (zh) 2021-02-19
KR20210025614A (ko) 2021-03-09
WO2020006374A3 (fr) 2020-02-20
US20210277113A1 (en) 2021-09-09
JP2021529520A (ja) 2021-11-04
EA202190138A1 (ru) 2021-05-27

Similar Documents

Publication Publication Date Title
US11634489B2 (en) Anti-TREM2 antibodies and methods of use thereof
US11319373B2 (en) Anti-SIRPA antibodies and methods of use thereof
KR102650524B1 (ko) 항-sirp-알파 항체 및 그의 사용 방법
US20210277113A1 (en) Anti-SIRP-Beta1 Antibodies and Methods of Use Thereof
AU2016316768A1 (en) Anti-Siglec-7 antibodies and methods of use thereof
AU2018310985A1 (en) Anti-CD33 antibodies and methods of use thereof
US20210317208A1 (en) Anti-cd33 antibodies and methods of use thereof
CN115003699A (zh) 抗trem2抗体的使用方法
AU2019282778A1 (en) Anti-Siglec-7 antibodies and methods of use thereof
EA044628B1 (ru) Анти-sirpa антитела и способы их применения
EA044134B1 (ru) Анти-trem2 антитела и способы их применения

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19744960

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 3099176

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020572914

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020026819

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019293589

Country of ref document: AU

Date of ref document: 20190628

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20217002521

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2019744960

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112020026819

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20201228